WO2017059425A1 - Waveguide-based detection system with scanning light source - Google Patents

Waveguide-based detection system with scanning light source Download PDF

Info

Publication number
WO2017059425A1
WO2017059425A1 PCT/US2016/055167 US2016055167W WO2017059425A1 WO 2017059425 A1 WO2017059425 A1 WO 2017059425A1 US 2016055167 W US2016055167 W US 2016055167W WO 2017059425 A1 WO2017059425 A1 WO 2017059425A1
Authority
WO
WIPO (PCT)
Prior art keywords
waveguides
optical
substrate
detection
excitation
Prior art date
Application number
PCT/US2016/055167
Other languages
French (fr)
Inventor
Reuven Duer
Yun-Pei CHANG
Ashutosh Shastry
Original Assignee
Ldip, Llc
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
Priority claimed from US15/245,062 external-priority patent/US20170082617A1/en
Application filed by Ldip, Llc filed Critical Ldip, Llc
Publication of WO2017059425A1 publication Critical patent/WO2017059425A1/en

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N21/00Investigating or analysing materials by the use of optical means, i.e. using sub-millimetre waves, infrared, visible or ultraviolet light
    • G01N21/62Systems in which the material investigated is excited whereby it emits light or causes a change in wavelength of the incident light
    • G01N21/63Systems in which the material investigated is excited whereby it emits light or causes a change in wavelength of the incident light optically excited
    • G01N21/64Fluorescence; Phosphorescence
    • G01N21/645Specially adapted constructive features of fluorimeters
    • G01N21/648Specially adapted constructive features of fluorimeters using evanescent coupling or surface plasmon coupling for the excitation of fluorescence
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N21/00Investigating or analysing materials by the use of optical means, i.e. using sub-millimetre waves, infrared, visible or ultraviolet light
    • G01N21/62Systems in which the material investigated is excited whereby it emits light or causes a change in wavelength of the incident light
    • G01N21/63Systems in which the material investigated is excited whereby it emits light or causes a change in wavelength of the incident light optically excited
    • G01N21/64Fluorescence; Phosphorescence
    • G01N21/645Specially adapted constructive features of fluorimeters
    • G01N21/6452Individual samples arranged in a regular 2D-array, e.g. multiwell plates
    • GPHYSICS
    • G02OPTICS
    • G02BOPTICAL ELEMENTS, SYSTEMS OR APPARATUS
    • G02B6/00Light guides; Structural details of arrangements comprising light guides and other optical elements, e.g. couplings
    • G02B6/24Coupling light guides
    • G02B6/26Optical coupling means
    • G02B6/35Optical coupling means having switching means
    • G02B6/3598Switching means directly located between an optoelectronic element and waveguides, including direct displacement of either the element or the waveguide, e.g. optical pulse generation
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N21/00Investigating or analysing materials by the use of optical means, i.e. using sub-millimetre waves, infrared, visible or ultraviolet light
    • G01N21/01Arrangements or apparatus for facilitating the optical investigation
    • G01N21/03Cuvette constructions
    • G01N2021/0346Capillary cells; Microcells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2201/00Features of devices classified in G01N21/00
    • G01N2201/08Optical fibres; light guides
    • GPHYSICS
    • G02OPTICS
    • G02BOPTICAL ELEMENTS, SYSTEMS OR APPARATUS
    • G02B6/00Light guides; Structural details of arrangements comprising light guides and other optical elements, e.g. couplings
    • G02B6/24Coupling light guides
    • G02B6/42Coupling light guides with opto-electronic elements
    • G02B6/4201Packages, e.g. shape, construction, internal or external details
    • G02B6/4204Packages, e.g. shape, construction, internal or external details the coupling comprising intermediate optical elements, e.g. lenses, holograms
    • G02B6/4206Optical features

Definitions

  • the present invention relates to methods and devices for generati ng optical pulses in one or more waveguides using a spatially scanning l ight source, and to detection systems, methods of use thereof and kits for detecti ng a biologically active analyte molecule.
  • Biological substance analysis methods based on optical means have risen in popularity in the last couple of decades. Common to all these methods is that chemical interactions between the bio- molecules produce changes that affect some measurable optical property, such as the emission spectrum, absorption spectrum or index of refraction.
  • the changes in the optical properties can occur in the analyte itself or through a mediator such as the surface on which the interaction takes place. These changes are then monitored using a beam of incoming light (usually laser light) which in turn changes the outgoing light spectrum (e.g., fluorescence), intensity (e.g., absorption), or phase (e.g., surface plasmon resonance and any kind of interferometric method).
  • a beam of incoming light usually laser light
  • intensity e.g., absorption
  • phase e.g., surface plasmon resonance and any kind of interferometric method.
  • microarray elements are spotted on top of a flat substrate chip usually made of glass, plastic or epoxy. Subsequently, the chip is scanned using confocal scanning systems where the exciting light and the resulting fluorescence light are both shined and collected from above and analyzed using a single photo-multiplier (PMT) detector.
  • PMT photo-multiplier
  • This arrangement suffers from several inherent limitations including a very short interaction length between the bio-sample and the light (usually a single mono-layer). This limits the signal strength and thus the SNR.
  • Another limitation is a high background or noise due to the fact that the back-reflected light and the emitted fluorescent light travel in the same direction.
  • a further limitation is high sensitivity to both the planarity and the position of the chip that need to be maintained in focus.
  • Stil l another limitation is slow operation due to the need to have large enough number of ' pixels' (scanned spots) within every sample and long enough integration time.
  • Yet another limitation is the need for a complicated optical and mechanical structure that
  • Another optical bio-analysis method is waveguide based bio-sensors. Bio-sensing based on waveguides has been around for a while. These biosensors can be divided into three main categories. The first involve slab waveguide fluorescence excitation with light collection from above or belo the chip. In this arrangement the bio-analyzed spots are located on the surface of a chip that contains a single slab- waveguide. Light is coupled into the waveguide using a lens or a grating that excites the entire chip with all its bio-analyzed spots simultaneously. The fluorescence is collected using an optical imaging system and a charge-coupled device (CCD) detector from above or underneath the chip.
  • CCD charge-coupled device
  • a second waveguide based bio-sensor utilizes an interferometric optical device.
  • channel waveguides are used together with interferometric devices such as Mach Zehnder interferometers (MZ1) or ring-resonators.
  • MZ1 Mach Zehnder interferometers
  • ring-resonators These sensitive interferometric devices sense the change in the index of refraction due to binding of the bio-molecules near a waveguide surface.
  • MZ1 Mach Zehnder interferometers
  • ring-resonators ring-resonators.
  • a third waveguide based bio-sensor utilizes surface plasmon resonance (SPR).
  • SPR surface plasmon resonance
  • a thin gold layer is deposited on top of a glass substrate.
  • the bio-analyzed sample on top of the gold induces changes in the refractive index above the gold layer, thus changing the resonant angle for generating surface plasmons along the gold layer.
  • the plasmon generation is detected as an enhanced peak in the reflected beam.
  • Examples of the SPR method are covered, for example, in U.S. Pat. No. 6,956,651 B2.
  • Other types of optical bio-sensors and array scanners exist such as described in U.S. Pat. No. 6,396,995 B l .
  • One aspect common to all of these waveguide based sensors is the need to initially couple light into the waveguide. Since all of these optical waveguides have miniature cross-sections ranging from 100 micrometers down to a fraction of a micrometer, the coupling of light into the waveguide involves specialized optics for focusing the light, fine mechanical alignment for accurately placing the light source relative to the waveguide, and specialized glues to bond all components in place without interfering with the light. This process adds in most of these cases a considerable cost and complexity to the entire system.
  • the l ight travels in the waveguide in the form of short pulses.
  • These pulses can be as short as a pico ( 1 0 ⁇ 12 ) second and as long as a few milli ( 10 -"1 ) seconds.
  • these pulses can be all of the same wavelength or can be a combination of many different wavelengths.
  • These pulses are generated by modulating one or more light sources which were initial ly coupled to the optical waveguide. If pulses at more than one wavelength are required, a combiner e.g., an Arrayed Waveguide Grating (AWG) must be added to the system.
  • AWG Arrayed Waveguide Grating
  • the optical waveguide may be part of a low-cost, consumable chip.
  • Waveguide-based optical detection systems are disclosed, for example, in U .S. Patent Nos. 7,951 ,583 B2, 8, 1 87.866 B2 and 8.288, 1 57 B2. each of which are hereby incorporated in their entirety by reference.
  • light needs to be coupled time after time to new chips.
  • the cost and complexity of the l ight coupl i ng technologies known in the art see, for example. U.S. Pat. Nos. 4.88 1 .789. 5.734.768. 5.600.744.
  • the optical signal generated on the sensing chip in all these applications is the result of one or more optical phenomena such as fluorescence, luminescence, absorption, scattering and optical phase change. Changes detected in the output light are indicative of the presence of the targets being detected. Nevertheless, the output light may also be affected by additional factors (e.g. temperature and aging) which may skew the results compromising the overall accuracy and precision of the detection system. Finding a method for normalizing the detected signal to overcome the effect of these uncontrol led factors is critical for maintaining the performance of the detection system.
  • the present invention provides methods and devices which allow the coupling of light into one or more optical waveguides in a simple and cost-effective manner.
  • the invention further provides methods and devices for simple coupling of multi-wavelength trains of pulses into an optical waveguide.
  • the invention further provides methods and devices for coupling correlated trains of pulses of one or more wavelengths into multiple optical waveguides.
  • the invention provides a method for generating an optical pulse in an optical waveguide, comprising providing an optical waveguide having an internal portion configured to carry an optical signal and a first end face in contact with said internal portion; providing a light beam; and spatially translating the light beam relative to the optical waveguide effective that the light beam transiently contacts the first end face of the optical waveguide, whereby an optical pulse is generated in the waveguide.
  • the invention provides a method for generating an optical pulse in an optical waveguide wherein the light beam has an optical mode, wherein the optical waveguide has an optical mode, and wherein while the light beam transiently contacts the optical waveguide, the light beam optical mode transiently overlaps the optical mode of the waveguide effective that l ight from the light beam passes into and within the optical waveguide.
  • the method further comprises providing a light source capable of emitting a light beam.
  • the light source is a laser.
  • the l ight source is a l ight emitting diode (LED).
  • the l ight beam is reflected prior to contacting the optical waveguide.
  • the l ight beam is refracted prior to contacting the optical waveguide.
  • the l ight source is movable, and spatial translation of the l ight beam emitted from the light source is effected by movement of the light source.
  • the movement of the l ight source is rotational, vertical, horizontal, transverse or longitudinal.
  • the waveguide is movable, and spatial translation of the l ight beam emitted from the l ight source is effected by movement of the waveguide.
  • the movement of the waveguide is rotational, vertical, horizontal, transverse or longitudi nal .
  • the light source is operably connected to an actuator.
  • the waveguide is operably connected to an actuator.
  • the movement of the actuator may be effected by electrical power, thermal power, magnetic power or even mechanical power (i.e., manually).
  • the actuator is a piezoelectric based motor, a step motor, an electrical motor, a magnetic actuator, a "memory metal" actuator, a solenoid, or a hydraulic actuator.
  • a light source is mounted on a piezoelectric bending actuator. Applying power to the actuator causes the actuator to move up and down, thereby scanning a line in space.
  • multiple light sources are mounted on a piezoelectric bending actuator. Every time the actuator scans its path, several optical pulses are injected into the optical waveguide. In some embodiments, the multiple light sources each emit light beams having a different wavelength. Every time the actuator scans its path, several optical pulses are injected into the optical waveguide, each at a different wavelength.
  • multiple optical waveguides are arranged along the scanning path of the one or more light sources.
  • a light source is mounted on the outer edge of a rotating disk, disposed effective that a light beam emitted from the light source is directed outwardly from the disk and transiently contacts an optical waveguide as the disk rotates.
  • multiple light sources are mounted on the rotating disk.
  • multiple optical waveguides are mounted around the rotating disk with their optical modes facing inward (toward the center of the disk). With every rotation of the disk, the optical mode of each and every light source wi ll overlap once with the optical mode of each and every optical waveguide. Therefore the total number of pulses generated in each optical waveguide will be equal to the total number of light sources mounted on the disk.
  • the light beams emitted from the l ight source are spatially translated.
  • the emitted l ight beams are spatially translated by a lens, a prism, a mirror, or a combination thereof.
  • the mirror is a stirring mirror.
  • a lens placed in front of the light source is translated in space, causing the light beam emitted by the light source to scan through space unti l its optical mode overlaps the optical mode of an optical waveguide.
  • a stirring mirror placed in front of the light source stirs the emitted light beam through space until its optical mode overlaps the optical mode of the optical waveguide.
  • the invention provides an apparatus for optical pulse generation in an optical waveguide, comprising a light source for emitting a l ight beam; an optical waveguide having a first end; and means for spatial ly translating a light beam from said light source such that the optical mode of the light source transiently contacts said the first end of the optical waveguide effective to provide an optical pulse in the waveguide.
  • the apparatus further comprises one or more additional light sources, and/or one or more additional optical waveguides.
  • the apparatus comprises multiple light sources each of which emits light beams having a different wavelength.
  • the means for spatially translating the light beam from the light source relative to the optical waveguide comprises a mechanism selected from a rotating disk, a motor, a solenoid, a hydraulic mechanism, a piezo-electric mechanism, and a "memory-metal" mechanism.
  • the light source is mounted on an outer edge of a rotatable disk, wherein said light source is disposed effective that a light beam emitted by said light source is directed outwardly of said rotatable disk, and that said light beam emitted by said light source is directed effective to transiently contact a first end of an optical waveguide as said disk rotates.
  • the light source is mounted on an actuator effective that a light beam emitted by said light source is directed effective to transiently contact a first end of an optical waveguide as said actuator moves.
  • the apparatus further comprises a scanning lens mounted on an actuator, wherein said scanning lens is disposed between the light source and the optical waveguide effective that movement of the actuator causes a light beam emitted by said light source to be directed by the lens effective to transiently contact a first end of an optical waveguide.
  • the apparatus further comprises a stirring mirror wherein said stirring mirror is disposed between the light source and the optical waveguide effective that movement of the stirring mirror causes a light beam emitted by said light source to be directed by the stirring mirror effective to transiently contact a first end of an optical waveguide.
  • the invention provides optical detection systems which utilize any of the above described methods or devices for coupling light into an optical waveguide.
  • the invention further provides the use of such detection systems for applications including but not limited to detection of a biological marker, detection of a chemical or biological warfare agent, detection or diagnosis of a viral or bacterial infectious disease, diagnosis of a genetic disorder or a cancer, detection of a protein-protein, protein-ligand, or protein-smal l molecule interaction, nucleic acid sequencing, and environmental monitoring of air, water, soil and food samples.
  • the present invention further provides detection systems and methods of use thereof including a scanning light source, a detector and a substrate comprising a plurality of waveguides and a plurality of optical sensing sites.
  • the light source is spatially translated relative to the substrate such that l ight emitted from the light source is coupled to and in optical communication with the waveguides of the substrate at some point along its scanning path.
  • the use of a scanning light source allows the coupl ing of l ight into the waveguides of the substrate in a simple and cost-effective manner.
  • the invention features a detection system and methods of use thereof including a scanning light source, a substrate comprising a plural ity of waveguides and a plurality of optical sensing sites in optical communication with one or more waveguide of the substrate, a detector that is coupled to and i n optical communication with the substrate, and means for spatial ly translating a l ight beam emitted from said scanning l ight source such that the l ight beam is coupled to and in optical communication with one or more waveguides of the substrate at some point along its scanning path.
  • the scanning light source is a chip comprising light generating elements.
  • the scanning light source chip further comprises waveguides.
  • the scanning l ight source is a chip that is further coupled to and in optical communication with an external light source.
  • the external light source is coupled to the scanning light source chip by optical fibers.
  • the scanning light source chip further includes waveguides.
  • the substrate comprises a plurality of substantial ly parallel excitation waveguides, and a plurality of substantially parallel collection waveguides, the excitation waveguides and collection waveguides crossing to form a two-dimensional array of intersection regions where an excitation waveguide and a collection waveguide cross and provide optical communication with the intersection region at each crossing; and a plurality of optica! sensing sites each in optical communication with an intersection region.
  • the system further comprises a scanning light source that is at some point along its scanning path coupled to and in optical communication with one or more of the excitation waveguides at a first edge of the substrate, and a detector that is coupled to and in optical communication with one or more of the collection waveguides at a second edge of the substrate.
  • two or more detectors are coupled to and in optical communication with one or more collection waveguides at various edges of the substrate.
  • the substrate comprises a plurality of substantially parallel waveguides, and a plural ity of optical sensing sites each in optical communication with a waveguide.
  • the system further comprises a scanning light source that is at some point along its scanning path coupled to and in optical communication with one or more of the waveguides at a first edge of the substrate, and a detector that is coupled to and in optical communication with said waveguides at the same or an opposite edge of the substrate.
  • the substrate comprises a plurality of in-coupl ing waveguides and a plurality of out-coupling waveguides, and a plurality of optical sensing sites each in optical communication with an in-coupling and an out-coupl ing waveguide.
  • the system further comprises a scanning light source that is at some point along its scanning path coupled to and in optical communication with one or more of the in-coupling waveguides at a first edge of the substrate, and a detector that is coupled to and in optical communication with one or more of the outcoupling waveguides of the substrate.
  • the scanning light source further comprises a detector, wherein at the point at which the light source is coupled to and in optical communication with one or more of the plurality of waveguides in optical communication with the optical sensing site, the detector is also coupled to and in optical communication with said one or more waveguides.
  • the substrate comprises a plurality of in-coupl ing waveguides and a pl ural ity of out-coupl ing waveguides, wherein at the point at which the l ight source is coupled to and in optical commun ication with one or more of the in-coupling waveguides, the detector is coupled to and in optical communication with one or more of the out-coupling waveguides.
  • the scanning light source is a chip comprising light generating elements and detector elements.
  • the scanning light source chip further comprises in-coupling and out-coupling waveguides.
  • the scanning light source chip further comprises at least one combiner.
  • the scanning light source is a chip that is further coupled to and in optical communication with an external light source and an external detector source.
  • the external light source is coupled to the scanning light source chip by optical fibers.
  • the scanni ng l ight source chip further includes incoupling and outcoupling waveguides.
  • the scanning l ight source chip further comprises at least one combiner.
  • the optical sensing site further comprises a sensor configured to transduce a first light wave generated by the l ight source in a waveguide, resulting in a second light wave in a different waveguide, the second l ight wave being detectable by the detector.
  • the optical sensing site further comprises a sensor configured to transduce a first light wave generated by the light source in a waveguide, resulting in a second light wave in the same waveguide, the second light wave being detectable by the detector.
  • the light source elements can provide variable wavelengths of light.
  • the light source is a broad-band source.
  • the light source is a tunable source.
  • the l ight source elements may be light emitting diodes (LEDs) or laser diodes (LDs).
  • the detector elements of the detector or of the scanning light source chip may be PIN diodes, avalanche photo-diodes, or a group of pixels which are part of a charge coupled device (CCD) array.
  • the detector is a silicon photodiode array.
  • the scanning l ight source is movable, and spatial translation of the light beam emitted from the l ight source is effected by movement of the scanning light source.
  • spatial translation of the light beam emitted from the l ight source is effected by movement of a component of the scanning light source, such as one or more mirrors, lenses, or prisms.
  • the substrate is movable, and, and spatial translation of the light beam emitted from the light source is effected by movement of the substrate.
  • the means for spatially translating the light beam from the light source relative to optical waveguides of the substrate comprises a mechanism selected from a rotating disk, a motor, a solenoid, a hydraul ic mechanism, a piezoelectric mechanism, and a memory-metal mechanism.
  • the scanning l ight source is operably connected to an actuator.
  • the movement of the actuator may be effected by electrical power, thermal power, magnetic power or even mechanical power (i.e.. manual ly).
  • the actuator is a piezoelectric based motor, a step motor, an electrical motor, a magnetic actuator, a memory metal actuator, a solenoid, or a hydraul ic actuator.
  • the actuator is a piezoelectric bending actuator.
  • the invention provides a detection method comprising delivering a sample suspected of containing a biologically active analyte molecule to be detected to an optical sensing site on the substrate of a detection system, and spatial ly translating a scanning light source to a point at which the light source is coupled to and in optical communication with one or more of a plurality of waveguides in optical communication with the optical sensing site, thereby generating a first light wave within said waveguide, wherein the first light wave is transducable by a sensor associated with the optical sensing site to a second l ight wave.
  • the method comprises detecting a measurable change in the second light wave using a detector in optical communication with the substrate, wherein a measurable change in the second l ight wave occurs when the sensor interacts with the biologically active analyte molecule.
  • the substrate comprises a plurality of substantially parallel excitation waveguides in optical communication with the optical sensing site, wherein the first light wave is transducable by a sensor associated with the optical sensing site to a second light wave carried in one or more of a plurality of substantially parallel collection waveguides in optical communication with the optical sensing site and crossing the excitation waveguides; and wherein a measurable change in the second light wave is detected using a detector in optical communication with the collection waveguides, wherein a measurable change in the second light wave occurs when the sensor interacts with the biologically active analyte molecule.
  • the substrate comprises a plurality of in-coupling and out-coupling waveguides in optical communication with the optical sensing site, and at the point at which the light source is coupled to and in optical communication with one or more of a plurality of in-coupling waveguides in optical communication with the optical sensing site, the detector is coupled to and in optical communication with one or more out-coupling waveguides.
  • the scanning l ight source further comprises a detector, and at the point at which the l ight source is coupled to and in optical communication with one or more waveguides in optical communication with the optical sensing site, the detector is also coupled to and in optical communication with said one or more waveguides.
  • the substrate comprises a plurality of in-coupling and out-coupling waveguides in optical communication with the optical sensing site, and the scanning light source further comprises a detector; and at the point at which the light source is coupled to and in optical communication with one or more of a pl urality of in-coupl ing waveguides in optical communication with the optical sensing site, the detector is coupled to and in optical communication with one or more out-coupl ing waveguides.
  • the biologically active analyte is selected from the group consisting of a nucleic acid, a protein, an antigen, an antibody, a microorganism, a gas, a chemical agent and a pollutant.
  • detecting a measurable change in the second light wave provides a diagnostic result.
  • a SNP is detected in the biological ly active analyte.
  • expression of a gene is detected upon detection of the biologically active analyte.
  • the method further comprises conducting a real-time PCR reaction at the optical sensing site.
  • the senor is adapted to support an immunoassay wherein the sensor interacting with the biologically active analyte comprises an outcome of an immunoassay.
  • the immunoassay supported is an enzyme-linked immunosorbent assay (ELISA).
  • the immunoassay supported is a fluorescent immunoassay.
  • the invention further provides the use of the detection systems for applications including but not limited to detection of biomarkers, detection of a chemical or biological warfare agent, detection or diagnosis of viral and bacterial infectious diseases, diagnosis of a genetic disorder or a cancer, detection of a protein-protein, protein-ligand, or protein-small molecule interaction, nucleic acid sequencing, and environmental monitoring of air, water, soi l and food samples.
  • a detection system for detecting a biologically active analyte molecule includes a substrate comprising one or more excitation waveguides, a plurality of collection waveguides, the one or more excitation waveguides and the plurality of collection waveguides crossing to form an array of intersection regions where an excitation waveguide and a collection waveguide cross and provide optical communication with the intersection region at each crossing, and a plurality of optical sensing sites each in optical communication with an intersection region; a scanning light source, wherein the scanning l ight source is at some point along its scanning path in optical communication with at least one of the one or more excitation waveguides; a detector that is in optical communication with one or more of the collection waveguides; and an actuator for spatially translating a light beam emitted from the scanning l ight source relative to the substrate such that the light beam is coupled to and in optical communication with at least one of the one or more excitation waveguides of the substrate at some point along its scanning path.
  • the one or more excitation waveguides is a single excitation waveguide.
  • both the one or more excitation waveguides and the plurality of collection waveguides exit the substrate at a first edge of the substrate.
  • the scanning l ight source is at some point along its scanning path in optical communication with one or more of the excitation waveguides at the first edge of the substrate; and the detector is in optical communication with one or more of the collection waveguides at the first edge of the substrate.
  • the one or more excitation waveguides are curved about 90 degrees.
  • a detection method includes delivering a sample suspected of containing a biological ly active analyte molecule to be detected to an optical sensing site on a substrate of a detection system, the substrate comprising one or more excitation waveguides and a plurality of col lection waveguides; spatially translating a scanning l ight source to a point at which the l ight source is in optical communication with at least one of the one or more excitation waveguides, wherein at least one of the one or more excitation waveguides is in optical communication with the optical sensing site, thereby generating a first light wave within said at least one of the one or more excitation waveguides, wherein the first light wave is transducable by a sensor associated with the optical sensing site to a second light wave carried in one or more of the plurality of collection waveguides in optical communication with the optical sensing site and crossing the one or more excitation waveguides; and detecting a measurable change in the second light wave using
  • the scanning l ight source further comprises a detector, and wherein at the point at which the light source is coupled to and in optical communication with one or more waveguides in optica! communication with the optical sensing site, the detector is also coupled to and in optical communication with said one or more waveguides.
  • the biologically active analyte is selected from the group consisting of a nucleic acid, a protein, an antigen, an antibody, a microorganism, a gas, a chemical agent and a pollutant.
  • the senor is adapted to support an immunoassay and wherein the sensor interacting with the biologically active analyte comprises an outcome of an immunoassay.
  • the one or more excitation waveguides is a single excitation waveguide.
  • both the one or more excitation waveguides and the plurality of collection waveguides exit the substrate at a first edge of the substrate.
  • the scanning light source is at some point along its scanning path in optical communication with one or more of the excitation waveguides at the first edge of the substrate; and the detector is in optical communication with one or more of the collection waveguides at the first edge of the substrate.
  • the one or more excitation waveguides are curved about 90 degrees.
  • the present invention relates to methods and systems of signal normalization, and more particularly to methods and systems of normal izing a signal generated in a waveguide based sensing chip used in bio-sensing applications.
  • a method of measuring a concentration of an analyte using a waveguide chip can include adding a known amount of one or more normalizing moieties to a sample; adding a first detection molecule labelled with a first fluorescent tag to the sample, the first detection molecule specific to the analyte; adding one or more secondary detection molecules, each secondary detection molecule label led with a distinct secondary fluorescent tag, to the sample, the one or more secondary detection molecules specific to the one or more normalizing moieties, wherein the first fluorescent tag and the one or more secondary fluorescent tags emit at different fl uorescence wavelengths: introducing the sample to an array of sensing sites on the waveguide chip, each sensing site of the array having a first capture molecule speci fic to the analyte and one or more secondary capture molecules specific to the one or more normal izing moieties, wherein the first capture molecule and the one or more secondary capture molecules are both immobil ized in each sensing site of the array at a
  • the step of normalizing the measured fluorescence emitted by the first detection molecule includes dividing the measured fluorescence emitted by the first detection molecule by the measured fluorescence emitted by the one or more secondary detection molecules.
  • the first detection molecule, the one or more secondary detection molecules, the first capture molecule, and the one or more secondary capture molecules are antibodies.
  • the step of normal izing the measured fluorescence emitted by the first detection molecule further includes generating one or more calibration curves from the measured fluorescence emitted by the one or more secondary detection molecules.
  • the one or more normalizing moieties includes at least a first normalizing moiety and a second normalizing moiety, wherein the amount of the first normalizing moiety added to the sample is greater than the amount of the second normalizing moiety added to the sample.
  • the step of normal izing the measured fluorescence emitted by the first detection molecule incl udes dividing the measured fluorescence emitted by the first detection molecule by a slope of the measured fluorescence emitted by the one or more secondary detection molecules.
  • the step of normalizing the measured fluorescence emitted by the first detection molecule includes dividing a slope of the measured fluorescence emitted by the first detection molecule by a slope of the measured fluorescence emitted by the one or more secondary detection molecules.
  • a method of measuring a concentration of an analyte using a waveguide chip can include adding a known amount of a first detection molecule label led with a first fluorescent tag to the sample, the first detection molecule specific to the analyte; introducing the sample to an array of sensing sites on the waveguide chip, each sensing site of the array having a first capture molecule speci fic to the analyte and a second capture molecule specific to the normal izing moiety, wherein the first capture molecule and the second capture molecule are both immobi l ized in each sensing site of the array at a predetermined ratio; introducing a first l ight pulse into one or more excitation waveguides on the waveguide chip at a first predetermined interval, the one or more excitation waveguides in optical communication with the array of sensing sites; measuring the fluorescence emitted by the first detection molecule through a plurality of collection waveguides in optical communication with the array of sensing sites
  • the step of normalizing the measured fluorescence emitted by the first detection molecule includes dividing the measured fluorescence emitted by the first detection molecule by the measured fluorescence emitted by the second detection molecule.
  • the step of normalizing the measured fluorescence emitted by the first detection molecule includes dividing the measured fluorescence emitted by the first detection molecule by an average of the measured fluorescence emitted by the second detection molecule.
  • the first predetermined interval is the same as the second
  • the first predetermined interval is different than the second predetermined interval.
  • the first light pulse and the second light pulse have the same wavelength.
  • a method of measuring a concentration of an analyte using a waveguide chip can incl ude adding a known amount of a first detection molecule labelled with a first fluorescent tag to the sample, the first detection molecule specific to the analyte; introducing the sample to an array of sensing sites on the waveguide chip, each sensing site of the array having an immobil ized capture molecule specific to the analyte; introducing a first light pulse into one or more excitation waveguides on the waveguide chip at a first predetermined interval, the one or more excitation waveguides in optical communication with the array of sensing sites; measuring the fl uorescence emitted by the first detection molecule through a plurality of collection waveguides in optical communication with the array of sensing sites; introducing a known amount of the analyte with a known amount of a second detection molecule labelled with a second fluorescent tag to the array of sensing sites, the second detection
  • the step of normalizing the measured fluorescence emitted by the first detection molecule includes dividing the measured fluorescence emitted by the first detection molecule by a slope of the measured fluorescence emitted by the second detection molecule.
  • the step of normalizing the measured fluorescence emitted by the first detection molecule includes dividing a slope of the measured fluorescence emitted by the first detection molecule by a slope of the measured fluorescence emitted by the second detection molecule.
  • the first light pulse and the second light pulse have the same wavelength.
  • FIG. 1 is a schematic drawing of an optical waveguide.
  • FIG. 2A is a schematic drawing illustrating one possible configuration of a light source for use in the invention.
  • FIG. 2B is a graph illustrating a typical light source optical mode.
  • FIG. 3A is a schematic drawing of an embodiment of a scanning-coupling system according to the invention.
  • FIG. 3B is a schematic drawing of a second embodiment of a scanning-coupling system according to the invention including multiple optical waveguides.
  • FIG. 3C is a schematic drawing of a third embodiment of a scanning-coupling system according to the invention including multiple light sources and multiple optical waveguides.
  • FIG. 4A is a schematic drawing of a fourth embodiment of a scanning-coupling system according to the invention including a rotating disk.
  • FIG. 4B is a schematic drawing of a fifth embodiment of a scanning-coupling system according to the invention including a rotating disk, multiple light sources and multiple optical waveguides.
  • FIG. 5 is a schematic drawing of a sixth embodiment of a scanning-coupl ing system according to the invention including a scanning lens.
  • FIG. 6 is a schematic drawing of a seventh embodiment of a scanning-coupl ing system according to the invention includi ng a stirring mirror.
  • FIG. 7A is a schematic of a detection system according to one embodiment of the invention including an active scanning light source chip, a substrate, a detector, and optical sensing sites.
  • FIG. 7B is a schematic of a detection system according to another embodiment of the invention including an active scanning l ight source/detector chip, a substrate and optical sensing sites.
  • FIG. 7C is a schematic of a detection system according to another embodiment of the invention including a light source, fibers, a passive scanning light source chip, a substrate, optical sensing sites and a detector.
  • FIG. 7D is a schematic of a detection system according to another embodiment of the invention including a light source, a detector, fibers, a passive light source/detector chip, a substrate and optical sensing sites.
  • FIG. 7E is a schematic side view of a detection system according to an embodiment of the invention including a scanning light source/detector chip, a piezoelectric bending actuator and a substrate.
  • FIG. 7F is a schematic of another embodiment of a substrate having a single excitation waveguide and a plurality of collection waveguides that exit the same side of the substrate.
  • FIG. 7G is a schematic of another embodiment of a substrate having a plurality of excitation waveguides and a pl ural ity of collection waveguides that exit the same side of the substrate.
  • FIG. 7H is a schematic of an embodiment of a substrate having excitation waveguides and collection waveguides and a loopback waveguide that exits the same side of the substrate.
  • FIG. 8 is a block diagram showing a representative example of a detection system of the invention in a housing as part of a working system.
  • FIG. 9A is a schematic of the substrate of the invention according to one embodiment including optical waveguides in conjunction with optical sensing sites and barriers.
  • FIG . 9B is a schematic of the substrate of the invention according to another embodiment including optical waveguides and combiners in conjunction with optical sensing sites and barriers.
  • FIG. 9C is a schematic cross section of the substrate of the invention according to one embodiment including an optical waveguide in conjunction with an optical sensing site.
  • FIG. 9D is a schematic of the substrate of the invention according to another embodiment including excitation and collection optical waveguides in conjunction with optical sensing sites and barriers.
  • FIG. 9E is a perspective view of the substrate of the embodiment of the invention shown in FIG. 9D including excitation and collection optical waveguides in conj unction with optical sensing sites.
  • FIG. 9F is a schematic of two cross sectional views (AA and BB) of the substrate shown in FIGS. 9D and 9E.
  • FIG . 9G is a schematic of a side view of the substrate of one embodiment of the invention in relation to a thermoelectric cooler.
  • FIG. 9H is a schematic of one embodiment of the substrate of the invention illustrating details of an optical sensing site including a heater and a thermistor.
  • FIG. 91 is a schematic of another embodiment of the substrate of the invention illustrating details of an optical sensing site including a heater and a thermistor.
  • FIG. 9J is a schematic of one embodiment of the substrate of the invention including reservoirs and micro channels in relation to optical sensing sites.
  • FIG. 9K is a schematic of another embodiment of the substrate of the invention including reservoirs and micro channels in relation to optical sensing sites.
  • FIG. 10A is a schematic of the substrate of the invention including excitation and collection optical waveguides in conjunction with optical sensing sites, barriers and funnels.
  • FIG. 10B is a schematic showing an enlarged view of substrate features according to an embodiment as shown in FIG. 10A.
  • FIG. I OC is a schematic of a cross-sectional view of the substrate according to one embodiment.
  • FIG. 1 1 A is a schematic of one embodiment of the substrate of the invention including excitation and collection optical waveguides in conjunction with optical sensing sites, barriers and branches.
  • FIG. 1 1 B is a schematic showing an enlarged view of substrate features according to an embodiment as shown in FIG. 1 1 A.
  • FIG. 1 1 C is a schematic of a cross-sectional view in a plane (AA) of the substrate according to one embodiment.
  • FIG. 1 1 D is a schematic of a cross-sectional view in a plane (BB) of the substrate according to one embodiment.
  • FIG. 12A is a schematic of a passive scanning light source chip of the invention according to one embodiment including optical in-coupling waveguides.
  • FIG. 12B is a schematic of a passive scanning light source/detector chip of the invention according to one embodiment including optical in-coupling and out-coupling waveguides and optical combiners.
  • FIG. 12C is a schematic of a passive scanning light source/detector chip of the invention according to another embodiment including optical in-coupling and out-coupling waveguides.
  • FIG. 13A is a schematic of a general substrate including typical layers and waveguides representative of those of the current invention.
  • FIG. 1 3 B is a photomicrograph image of waveguides representative of those of the invention and a silica layer.
  • F IG. 13C is a perspective view of waveguides and associated substrate layers.
  • FIG. 14A is a schematic of an active scanning l ight source chip of the invention according to one embodiment including l ight source elements and optical in-coupl ing waveguides.
  • FIG. 14B is a schematic of an active scanning l ight source/detector chip of the invention according to one embodiment including light source elements, detector elements, optical in-coupling and out-coupl ing waveguides and optical combiners.
  • FIG. 15 is a block diagram showing a representative example logic device in communication with an apparatus for use with the detection system of the invention.
  • FIG. 16 is a block diagram showing a representative example of a kit.
  • FIGS. 17A-D are schematics illustrating a representative manufacturing process for the substrate and waveguides of the invention.
  • FIG. 18 is a flow chart showing a representative manufacturing process for the substrate.
  • FIG. 19 is a plot of data for fluorescently labeled ovalbumin binding to immobilized antibodies specific for ovalbumin.
  • FIG. 20 is a plot of data for incorporation of Cy5.5-labeled cytosines into a DNA molecule during a primer extension reaction as detected using one embodiment of a detection system of the invention.
  • FIG. 21 is a graph showing a real-time detection of Clostridium difficile toxin A in bovine serum on a 10-channel chip using a detection system of the invention.
  • FIG. 22 is a graph showing a standard curve for Clostridium difficile toxin A as measured on a series of ten 10-channel chips using a detection system of the invention.
  • FIG. 23 is a flow chart illustrating a procedure for aligning a cartridge in a reader.
  • FIG. 24 is a flow chart illustrating a procedure for immobilizing antibodies to a substrate.
  • FIGS. 25A-25D illustrate an array of droppers that can dispense droplets of antibodies onto a substrate.
  • the present invention provides methods and devices for coupling light of one or more wavelengths into one or more optical waveguides in a simple and cost-effective manner.
  • Apparatus, methods, and kits for optical detection, using a detection system including a scanning light source, a detector, a substrate, and a plurality of waveguides and optical sensing sites are also provided.
  • One substrate of the present system includes a plurality of substantially parallel excitation waveguides and a plurality of substantially parallel collection waveguides. The excitation waveguides and collection waveguides cross to form an intersection region and a two-dimensional array.
  • Other substrates of the present system include a plurality of substantially parallel waveguides and a plural ity of sensing sites.
  • the optical sensing sites include a sensor and are in optical communication with one or more waveguides. Detection of a variety of environmental and biological samples can be achieved using the apparatus, methods and kits described herein. The general theoretical principles of lightwave guiding and evanescent field fluorescence excitation apply to the embodiments disclosed herein.
  • biologically active analyte when used herein means any substance which can affect any physical or biochemical properties of a biological organism, including but not limited to viruses, bacteria, fungi, plants, animals, and humans.
  • biologically active analyte according to the present invention includes without limitation drugs, prodrugs, pharmaceutical agents, drug metabolites, biomarkers such as expressed proteins and cell markers, antibodies, serum proteins, cholesterol, polysaccharides, nucleic acids, biological analytes, genes, proteins, or hormones, or any combination thereof.
  • a biologically active analyte can further include a natural or man-made substance including but not limited to a gas, a chemical agent or a pollutant, or a combination thereof (e.g., from an environmental source).
  • the biologically active analytes can be polypeptide, glycoprotein, polysaccharide, lipid, nucleic acid, or a combination thereof.
  • biomarkers associated with a particular disease or with a specific disease stage are biomarkers associated with a particular disease or with a specific disease stage.
  • Such biologically active analytes include but are not limited to those associated with autoimmune diseases, obesity, hypertension, diabetes, neuronal and/or muscular degenerative diseases, cardiac diseases, endocrine disorders, and any combinations thereof.
  • biomarkers that are present in varying abundance in one or more of the body tissues including heart, liver, prostate, lung, kidney, bone marrow, blood, skin, bladder, brain, muscles, nerves, and selected tissues that are affected by various disease, such as different types of cancer (malignant or non-metastatic), autoimmune diseases, inflammatory or degenerative diseases.
  • body tissues including heart, liver, prostate, lung, kidney, bone marrow, blood, skin, bladder, brain, muscles, nerves, and selected tissues that are affected by various disease, such as different types of cancer (malignant or non-metastatic), autoimmune diseases, inflammatory or degenerative diseases.
  • microorganisms include but are not limited to bacterium, virus, fungus and protozoa.
  • Biologically active analytes that can be detected by the subject method also include blood-born pathogens selected from a non-limiting group that consists of Staphylococcus epiclermidis, Escherichia
  • MSRA methici llin-resistant Staphylococcus aureus
  • Staphylococcus aureus Staphylococcus hominis
  • Enterococcus faecalis Pseudomonas aeruginosa
  • Staphylococcus capitis Staphylococcus warneri
  • Klebsiella pneumoniae Haemophilus influnzae
  • Staphylococcus simulcms Streptococcus pneumoniae and Candida albicans.
  • Biologically active analytes that can be detected by the subject device and methods also encompass a variety of sexually transmitted diseases selected from the fol lowing: gonorrhea (Neisseria gorrhoeae), syphil is ⁇ Treponena pallidum), chlamydia (Chlamydia iracomi/is), nongonococcal urethritis ( Ureaplasma urealyticum), yeast infection (Candida albicans), chancroid (Haemophilus ducreyi), trichomoniasis (Trichomonas vaginalis), genital herpes (HSV type I and II), H IV I, HI V II and hepatitis A, B, C, G, as well as hepatitis caused by TTV.
  • gonorrhea Neisseria gorrhoeae
  • syphil is ⁇ Treponena pallidum
  • chlamydia
  • Additional biologically active analytes that can be detected by the subject apparatus and methods encompass a diversity of respiratory pathogens including but not limited to Pseudomonas aeruginosa, methici llin-resistant Staphylococcus aureus(MSRA), Klebsiella pneumoniae, Haemophilis influenzae, Staphylococcus aureus, Stenotrophomonas maltophilia, Haemophilis parainfluenzae, Escherichia coli, Enterococcus faecalis, Serratia marcescens, Haemophilis par ahaemolyticus,
  • MSRA methici llin-resistant Staphylococcus aureus
  • MSRA methici llin-resistant Staphylococcus aureus
  • Klebsiella pneumoniae Haemophilis influenzae
  • Staphylococcus aureus Stenotrophomonas maltophilia
  • Enterococcus cloacae Candida albicans, Moraxiella catarrhalis, Streptococcus pneumoniae, Citrobacter penindii, Enterococcus faecium, Klebsiella oxytoca, Pseudomonas fluorsecens, Neisseria meningitidis, Streptococcus pyogenes, Pneumocystis carinii, Klebsiella pneumoniae, Legionella pneumophila, Mycoplasma pneumoniae, and Mycobacterium tuberculosis.
  • Theophylline CRP, C MB, PSA, Myoglobin, CA 125, Progesterone, TxB2, 6-keto-PGF-l -alpha, and Theophylline, Estradiol, Lutenizing hormone, High sensitivity CRP, Triglycerides, Tryptase, Low density l ipoprotein Cholesterol, High density lipoprotein Cholesterol, Cholesterol, IGFR.
  • liver markers include without limitation LDH, (LD5), (ALT), Arginase 1 (liver type), Alphafetoprotein (AFP), Alkaline phosphatase, Alanine aminotransferase, Lactate dehydrogenase, and Bilirubin.
  • kidney markers include without limitation TNFa Receptor, Cystatin C, Lipocalin-type urinary prostaglandin D, synthatase (LPGDS), Hepatocyte growth factor receptor, Polycystin 2, Polycystin 1 , Fibrocystin, Uromodulin, Alanine, aminopeptidase, N-acetyl-B-D- glucosaminidase, Albumin, and Retinol-binding protein (RBP).
  • TNFa Receptor include without limitation TNFa Receptor, Cystatin C, Lipocalin-type urinary prostaglandin D, synthatase (LPGDS), Hepatocyte growth factor receptor, Polycystin 2, Polycystin 1 , Fibrocystin, Uromodulin, Alanine, aminopeptidase, N-acetyl-B-D- glucosaminidase, Albumin, and Retinol-binding protein (RBP).
  • Exemplary heart markers include without limitation Troponin I (Tnl), Troponin T (TnT), CK, CKMB, Myoglobin, Fatty acid binding protein (FABP), CRP, D-dimer, S- 100 protein, BNP, NT- pro BNP, PAPP-A, Myeloperoxidase (MPO), Glycogen phosphorylase isoenzyme BB (GPBB), Thrombin Activatable Fibrinolysis Inhibitor (TAFI), Fibrinogen, Ischemia modified albumin (IMA), Cardiotrophin- 1 , and LC-1 (Myosin Light Chain-I).
  • Tnl Troponin I
  • TnT Troponin T
  • CK Troponin T
  • CKMB Myoglobin
  • CRP D-dimer
  • S- 100 protein BNP, NT- pro BNP, PAPP-A, Myeloperoxidase (MPO), Glycogen phosphorylase is
  • pancreas markers include without limitation Amylase, Pancreatitis-Associated protein (PAP-1 ), and Regenerated proteins (REG).
  • PAP-1 Pancreatitis-Associated protein
  • REG Regenerated proteins
  • Exemplary muscle tissue markers include without limitation Myostatin.
  • Exemplary blood markers include without limitation Erythopoeitin (EPO).
  • EPO Erythopoeitin
  • Exemplary bone markers include without limitation, Cross-l inked N-telopeptides of bone type I col lagen (NTx), Carboxyterminal cross-linking telopeptide of bone collagen, Lysyl-pyridinol ine (deoxypyridinol ine), Pyridinol ine, Tartrate-resistant acid phosphatase, Procollagen type I C propeptide,
  • Procollagen type I N propeptide Osteocalcin (bone gla-protein), Alkal ine phosphatase, Cathepsin K,
  • CO P Cartilage Oligomeric Matrix Protein
  • Osteocrin Osteoprotegerin
  • OPG Osteoprotegerin
  • RA L sRANK
  • TRAP 5 TRACP 5
  • OSF-1 Osteoblast Specific Factor 1
  • OSF-2 Osteoblast Specific Factor 2
  • OSF-2 Osteoblast Specific Factor 2
  • markers according to the present invention are disease specific.
  • Exemplary cancer markers include without limitation PSA (total prostate specific antigen), Creatinine, Prostatic acid phosphatase, PSA complexes, Prostrate-specific gene- 1 , CA 12-5, Carcinoembryonic
  • CEA Alpha feto protein
  • hCG Human chorionic gonadotropin
  • Inhibin CAA Ovarian C 1 824, CA 27.29, CA 15-3, CAA Breast C 1924, Her-2, Pancreatic, CA 19-9, Carcinoembryonic Antigen, CAA pancreatic, Neuron-specific enolase, Angiostatin.
  • DcR3 Soluble decoy receptor 3
  • Endostatin Ep- CAM ( K- 1 )
  • Free Immunoglobulin Light Chain Kappa Free Immunoglobulin Light Chain Lambda
  • Herstatin Chromogranin A
  • Adrenomedullin Integrin
  • Epidermal growth factor receptor Epidermal growth factor receptor-Tyrosine kinase
  • Pro-adrenomedullin N-terminal 20 peptide Vascular endothelial growth factor, Vascular endothelial growth factor receptor, Stem cell factor receptor, c-kit/KDR, KDR, and Midkine.
  • infectious disease markers include without limitation Viremia, Bacteremia, Sepsis, PMN Elastase, PMN elastase/al -Pl complex, Surfactant Protein D (SP-D), HBVc antigen, HBVs antigen, Anti-HBVc, Anti-HIV, T-suppressor cell antigen, T-cell antigen ratio, T-helper cell antigen, Anti-HCV, Pyrogens, p24 antigen and Muramyldipeptide.
  • SP-D Surfactant Protein D
  • HBVc antigen HBVs antigen
  • Anti-HBVc Anti-HIV
  • T-suppressor cell antigen T-cell antigen ratio
  • T-helper cell antigen Anti-HCV
  • Pyrogens Pyrogens
  • p24 antigen Muramyldipeptide.
  • Exemplary diabetes markers include without limitation C-Peptide, Hemoglobin Ale,
  • Glycated albumin Advanced glycosylation end products (AGEs), 1 ,5-anhydroglucitol, Gastric Inhibitory Polypeptide, Glucose, Hemoglobin, ANGPTL3 and ANGPTL 4.
  • AGEs Advanced glycosylation end products
  • 1 ,5-anhydroglucitol Gastric Inhibitory Polypeptide
  • Glucose Glucose
  • Hemoglobin ANGPTL3 and ANGPTL 4.
  • Exemplary inflammation markers include without limitation Rheumatoid factor (RF),
  • ANA Antinuclear Antibody
  • CRP C-reactive protein
  • Clara Cell Protein Clara Cell Protein
  • Exemplary allergy markers include without limitation Total IgE and Specific IgE.
  • autism markers include without l imitation Ceruloplasmin, Metalothioneine, Zinc, Copper, B6, B 12, Glutathione, Alkaline phosphatase, and Activation of apo-alkaline phosphatase.
  • Exemplary coagulation disorders markers include without limitation b-Thromboglobulin,
  • a marker may be therapy specific.
  • COX inhibitors include without limitation TxB2 (Cox- 1 ), 6-keto-PGF-l -alpha (Cox 2) and 1 1 -Dehydro-TxB- l a (Cox- 1 ).
  • Exemplary geriatric markers include without limitation, Neuron-specific enolase, GFAP and S 100B.
  • Exemplary markers of nutritional status incl ude without limitation Prealbumin, Albumin, Retinol-binding protein (RBP), Transferrin, Acylation-Stimulating Protein (ASP), Adiponectin, Agouti- Related Protein (AgRP), Angiopoietin-like Protein 4 (ANGPTL4, FIAF), C-peptide, AFABP (Adipocyte Fatty Acid Binding Protein, FAB P4). Acylation-Stimulating Protein ( ASP). EFA BP ( Epidermal Fatty Acid Binding Protein, FABP5), Glicentin, Gl ucagon. G lucagon-Like Peptide- 1 . Gl ucagon-Like Peptide-2, Ghrelin, Insulin, Leptin, Leptin Receptor, PYY, RELMs, Resistin, and sTfR (soluble Transferrin Receptor).
  • ASP Acylation-Stimulating Protein
  • FABP Epidermal Fatty Acid
  • Exemplary markers of lipid metabolism include without limitation Apo-lipoproteins (several), Apo-A l , Apo-B, Apo-C-Cll, Apo-D and Apo-E.
  • Exemplary coagulation status markers include without limitation Factor 1 : Fibrinogen, Factor II : Prothrombin, Factor II I : Tissue factor, Factor IV: Calcium, Factor V: Proaccelerin, Factor VI, Factor VII: Proconvertin, Factor VIII :, Anti-hemolytic factor, Factor IX : Christmas factor, Factor X: Stuart-Prower factor, Factor XI : Plasma thromboplastin antecedent, Factor XI I : Hageman factor, Factor XI I I: Fibrin-stabilizing factor, Prekallikrein, High-molecular-weight kininogen, Protein C, Protein S, D- dimer, Tissue plasminogen activator, Plasminogen, a2-Antiplasmin and Plasminogen activator inhibitor 1 (PAI 1 ).
  • Factor 1 Fibrinogen
  • Factor II Prothrombin
  • Factor II I Tissue factor
  • Factor IV Calcium
  • Factor V Proacceler
  • Exemplary monoclonal antibody markers include those for EGFR, ErbB2, and IGF 1 R.
  • Exemplary tyrosine kinase inhibitor markers incl ude without limitation Abl, Kit, PDGFR, Src, ErbB2, ErbB 4, EGFR, EphB, VEGFRl -4, PDGFRb, FLt3, FGFR, PKC, Met, Tie2, RAF, and TrkA.
  • Exemplary serine/threonine kinase inhibitor markers include without limitation AKT, Aurora A/B/B, CDK, CDK (pan), CD l -2, VEGFR2, PDGFRb, CD 4/6, MEK 1 -2, mTOR, and PKC- beta.
  • GPCR target markers include without limitation Histamine Receptors, Serotonin Receptors, Angiotensin Receptors, Adrenoreceptors, Muscarinic Acetylcholine Receptors, GnRFl Receptors, Dopamine Receptors, Prostaglandin Receptors, and ADP Receptors.
  • a "therapeutic agent” is intended to include any substances that have therapeutic utility and/or potential. Such substances include but are not limited to biological or chemical compounds such as simple or complex organic or inorganic molecules, peptides, proteins (e.g. antibodies) or polynucleotides (e.g. anti-sense).
  • biological or chemical compounds such as simple or complex organic or inorganic molecules, peptides, proteins (e.g. antibodies) or polynucleotides (e.g. anti-sense).
  • a vast array of compounds can be synthesized, for example, polymers, such as polypeptides and polynucleotides, and synthetic organic compounds based on various core structures, and these are also included in the term "therapeutic agent".
  • various natural sources can provide compounds for screening, such as plant or animal extracts, and the like. It should be understood, although not always explicitly stated, that the agent is used alone or in combination with another agent, having the same or different biological activity as the agents identified by the inventive screen. The agents and methods also are intended
  • Pharmacodynamic (PD) parameters include without l imitation physical parameters such as temperature, heart rate/pulse, blood pressure, and respiratory rate, and biomarkers such as proteins, cells, and cell markers. Biomarkers could be indicative of disease or could be a result of the action of a drug.
  • Pharmacokinetic (PK) parameters according to the present invention include without l imitation drug and drug metabolite concentration. Identifying and quantifying the PK parameters rapidly from a sample vol ume is extremely desirable for proper safety and efficacy of drugs. If the drug and metabol ite concentrations are outside a desired range and/or unexpected metabol ites are generated due to an unexpected reaction to the drug, immediate action may be necessary to ensure the safety of the patient. Similarly, if any of the PD parameters fall outside the desired range during a treatment regime, immediate action may have to be taken as well.
  • physical parameter data is stored in or compared to stored profiles of physical parameter data in a bioinformatics system which may be on an external device incorporating pharmacogenomic and pharmacokinetic data i nto its models for the determination of toxicity and dosing. Not only does this generate data for clinical trials years prior to current processes but also enables the elimination of current disparities between apparent efficacy and actual toxicity of drugs through real-time continuous monitoring. During the go/no go decision process in clinical studies, large scale comparative population studies can be conducted with the data stored on the database. This compilation of data and real-time monitoring allows more patients to enter clinical trials in a safe fashion earlier than currently allowed.
  • biomarkers discovered in human tissue studies can be targeted by the detection system for improved accuracy in determining drug pathways and efficacy in cancer studies.
  • nucleic acid when used herein refers to deoxyribonucleotides
  • deoxyribonucleosides, ribonucleosides or ribonucleotides and polymers thereof in either single- or double-stranded form encompasses nucleic acids containing known analogues of natural nucleotides which have similar binding properties as the reference nucleic acid and are metabolized in a manner similar to naturally occurring nucleotides. Unless specifically limited otherwise, the term also refers oligonucleotide analogs including PNA (peptidonucleic acid), analogs of DNA used in antisense technology (phosphorothioates, phosphoroamidates, and the like). Unless otherwise indicated, a particular nucleic acid sequence also impl icitly encompasses conservatively modified variants thereof (including but not limited to, degenerate codon substitutions) and
  • degenerate codon substitutions may be achieved by generating sequences in which the third position of one or more selected (or all) codons is substituted with mixed-base and/or deoxyinosine residues (Batzer et al., Nucleic Acid Res. 19:5081 ( 1991 ); Ohtsuka et al., J. Biol. Chem. 260:2605-2608 ( 1 985); and Rossolini et al ., Mol. Cell. Probes 8:91 -98 ( 1994)).
  • microorganism' when used herein refers to bacteria, actinomycetales, cyanobacteria (unicel lular algae), fungi, protozoa, animal cells or plant cells or viruses. Examples of microorganisms include but are not limited to pathogens.
  • polypeptide peptide
  • protein protein
  • a description directed to a polypeptide applies equally to a description of a peptide and a description of a protein, and vice versa.
  • the terms apply to naturally occurring amino acid polymers as well as amino acid polymers in which one or more amino acid residues is a non-natural amino acid.
  • the terms encompass ami no acid chains of any length, including ful l length proteins (i.e., antigens), wherein the amino acid residues are linked by covalent peptide bonds.
  • proteins that contain multiple polypeptide chains that associate through covalent and/or non-covalent interactions are also encompassed by "protein, " as used herein.
  • polymorphism refers to the occurrence of two or more genetically determined alternative sequences or alleles in a population.
  • a polymorphic marker or site is the locus at which divergence occurs. Preferred markers have at least two alleles, each occurring at a frequency of greater than 1 %, and more preferably greater than 10% or 20% of a selected population.
  • a polymorphism may comprise one or more base changes, an insertion, a repeat, or a deletion.
  • a polymorphic locus may be as small as one base pair.
  • Polymorphic markers include restriction fragment length polymorphisms, variable number of tandem repeats (VNTR's), hypervariable regions, minisatellites, dinucleotide repeats, trinucleotide repeats, tetranucleotide repeats, simple sequence repeats, and insertion elements such as Alu.
  • the first identified allelic form is arbitrarily designated as the reference form and other allelic forms are designated as alternative or variant alleles.
  • the allelic form occurring most frequently in a selected population is sometimes referred to as the wildtype form. Diploid organisms may be homozygous or heterozygous for al lelic forms.
  • a diallelic polymorphism has two forms.
  • a triallelic polymorphism has three forms.
  • a single nucleotide polymorphism occurs at a polymorphic site occupied by a single nucleotide, which is the site of variation between allelic sequences.
  • the site is usually preceded by and followed by highly conserved sequences of the allele (e.g., sequences that vary in less than 1 /100 or 1 /1000 members of the populations).
  • a single nucleotide polymorphism usually arises due to substitution of one nucleotide for another at the polymorphic site.
  • a transition is the replacement of one purine by another purine or one pyrimidine by another pyrimidine.
  • a transversion is the replacement of a purine by a pyrimidine or vice versa.
  • Single nucleotide polymorphisms can also arise from a deletion of a nucleotide or an insertion of a nucleotide relative to a reference al lele.
  • the term "individual” when used herein is not limited to a human being, but may also include other organisms including but not limited to mammals, plants, bacteria or cells derived from any of the above.
  • aspects of the invention may include one or more of the fol lowing advantageous features.
  • Dense and accurate integration of optical manipulating elements can be achieved using planar lightwave circuit technology.
  • Appl ications for planar lightwave circuits as described herein include new drug discovery and development, disease research, biomarkers discovery, detection of a chemical or biological warfare agent, environmental monitoring, SNP association studies including toxicology and disease susceptibi l ity, and diagnostics incl uding identifying patients predisposed to diseases and identifying patients with particular drug sensitivity.
  • optical coupling from one clement to another (e.g. a light emitter to an opt ical waveguide) occurs whenever there is some overlap between the “optical modes " of the two elements.
  • the "optical mode” of an element represents the spatial and temporal behavior of the light emitted from or accepted by that element.
  • one or more light sources or alternatively, their emitted light beams are spatially translated relative to one or more optical waveguides with the optical modes of the light sources scanning through space to overlap at some arbitrary time and arbitrary place with some of the optical modes of one or more optical waveguides.
  • light is coupled from the overlapping optical mode of a light source to the overlapping optical mode of an optical waveguide, thereby generating a light pulse within the optical waveguide, which pulse has a duration equal to the overlapping time and a wavelength equal to that of the overlapping light source.
  • spatially translated relative to the optical waveguide is meant that either the light sources are physically translated through space, or their emitted light beams are translated using optical means (e.g. lenses, prisms, mirrors, etc.) or the optical waveguides are physically translated through space, until the overlap of the optical modes occurs.
  • optical means e.g. lenses, prisms, mirrors, etc.
  • a general optical waveguide 101 is depicted in FIG. 1 . It consists of two main regions, the core region 1 1 1 and the cladding region 1 10. Light is confined and propagates along a waveguide in the core region. This confinement is most commonly achieved by designing the core region to have an index of refraction higher than that of the cladding region. Thus light 'trying to escape' the core region experiences 'total internal reflection' and remains trapped in the core region. Only light having certain properties (e.g., propagation angle) is trapped within the core region. Every optical waveguide has a discrete set of "modes" having those properties and thus able to propagate along the waveguide. These are referred to as the "optical modes" of the waveguide 1 12.
  • a representative light source 202 is schematically shown in FIG. 2A.
  • the light source consists of a laser chip 223 and the electrical leads 221 for driving it.
  • lasers are the most common source for coupling light into a waveguide, in some cases a Light Emitting Diode (LED) may be used.
  • a lens 222 in the drawing represents a variety of optical components that can be used to manipulate (i.e. collimate, focus, filter, deflect, etc.) the emitted light. Al l these components can be assembled in a compact way on a single or on multiple substrates 224.
  • Such a light source is connected to its driving electronics which control the way it operates to generate a continuous wave (CW) of light or pulses.
  • the l ight source generates l ight in one or in several different wavelengths. In some cases, the wavelength of the l ight emitted by the light source can be tuned by changing its temperature or by placing a filter in front of the light source to pick one wavelength out of the emitted spectrum.
  • the light emitted by the light source and passing through the lens (and/or other optical components) has a certain intensity distribution and propagation angles, referred to herein as the light source "optical mode".
  • FIG. 2B shows an example of a l ight source optical mode 21 3, its intensity distribution 21 4 at a given distance (Z).
  • a pulse of light from the light source is coupled to the optical waveguide by moving the one relative to the other through a point where there is some overlap between the optical modes of the two, resulting in the generation of an optical pulse within the optical waveguide.
  • Repeating the same process with the same light source and optical waveguide generates a train of identical light pulses.
  • Repeating the same process with different light sources generates a train of pulses differing in wavelength, and/or duration and/or any other property of the light emitted by the light sources (e.g., intensity, temporal coherence, spatial coherence, or amplitude modulation).
  • Repeating the scanning process with multiple light sources and multiple optical waveguides generates correlated trains of pulses in all or some of the optical waveguides.
  • the generated pulse will be a convolution in space of the two optical modes of the light source and the optical waveguide. This convolution is affected by both of the optical modes and their spatial and temporal overlap. The resulting pulse is also affected by the size of the two beams. The scanning speed also affects the generated pulse through the temporal dependence of the moving optical mode. If the waveguide has more than one mode the resulting pulse will be a sum of all overlaps.
  • the pulse duration may be more simply calculated by taking the effective beam diameter of the light source and that of the waveguide.
  • the approximate pulse duration is the sum of these two beam diameters divided by the scanning amplitude, multiplied by the scanning period.
  • the relative movement of the one or more light sources or their emitted light beams with respect to the one or more optical waveguides may include mechanisms such as, for example, a rotating disk, a motor, a solenoid, a hydraulic mechanism, a piezo-electric mechanism, and a "memory-metal" mechanism.
  • the relative motion may be generated through simple manually driven actuators.
  • Either the light sources or the optical waveguides may be physically translated through space.
  • the light sources may be stationary while the light beams emitted from the light sources are translated using optical means (e.g. lenses, prisms, mirrors, etc.). Spatial translation of the optical means may be achieved using any of the above described means.
  • the light source 302 is mounted on a piezoelectric bending actuator 305 as shown in FIG. 3A.
  • the scanning-coupling system 300 further comprises an optical waveguide 301 .
  • the piezoelectric bending actuator driver (not shown) interfaces to the actuator through electrical wires 306.
  • the driver generates the required voltage to run the actuator. This voltage may be in the form of a 'Saw-Tooth' wave, or a ' Sine ' wave or a ' Square' wave or any other form of electrical wave that would cause the piezoelectric bending actuator to bend up and/or down, thus moving the light source relative to the optical waveguide.
  • the duration of the pulse is equal to the overlap time of the two optical modes.
  • the duration of the pulse may be controlled either by changing one of the optical modes or by adjusting the scanning speed.
  • the optical mode of the light source can be controlled by, for example, moving the lens in front of the emitter (see FIG. 2A) away or closer, thus expanding or converging the emitted light beam (i.e. the optical mode).
  • the width of the overlap of the two optical modes will be a convolution of the two, resulting in a Gaussian shaped optical mode of ⁇ 10-microns in width.
  • the piezoelectric bending actuator is driven with a periodic ' Saw-Tooth' wave having a frequency of 100 Hz with a scanning amplitude of 300 microns, the result is a 167 micro second pulse generated in the optical waveguide once every 5 milliseconds.
  • the piezoelectric bending actuator has limitations on its scanning speed.
  • the components of the scanning-coupling system of the invention are therefore selected or designed to be able to meet the required scanning speed for a speci fic application.
  • the current invention may be particularly advantageous in applications where the duration of the light pulses involved is of the order of a micro ( 10 ⁇ 6 ) second or longer.
  • FIG. 3 B A further embodiment of the invention is shown in FIG. 3 B, where the scanning-coupl ing system 300 consists of the piezoelectric bending actuator 305 and its electrical wires 306, the light source 302 and multiple optical waveguides 301 .
  • the scanning light source will now generate trains of pulses in some or all of the optical waveguides based upon the chosen scanning path.
  • the scanning-coupling system 300 comprises multiple light sources 302, multiple optical waveguides 301 , a piezoelectric bending actuator 305 and its electrical wires 306.
  • the properties of the different light sources for example, their wavelength or optical mode.
  • the light source 402 is mounted on a rotating disk 403 to create a scanning-coupl ing system 400.
  • the system further comprises an optical waveguide 401 .
  • the rotating disk is driven by an electronic driver (not shown) causing the light source optical mode to scan the periphery of the disk through a point where it overlaps the optical mode of an optical waveguide.
  • a light pulse is generated in the optical waveguide, the duration of which is equal to the duration of the overlap.
  • the pulse duration is controlled by the shape of the two optical modes and the rotating speed of the disk.
  • FIG. 4B A further embodiment of the scanning-coupl ing system 400 is shown in FIG. 4B.
  • Multiple light sources 402 are mounted on a disk 403 facing outwards, with multiple optical
  • the time between two adjacent pulses in the same opt ical waveguide is controlled by the spacing between the l ight sources, as well as the rotation speed of the disk. There is also a well defined time correlation between the trains of pulses in all the optical waveguides.
  • the scanning-coupl ing system 500 consists of a piezoelectric bending actuator 505 and its electrical wires 506, mounted with a scanning lens 507.
  • the scanning of the light beam emitted by the light source is achieved by moving the lens in front of the light source.
  • the lens is disposed between the light source and the optical waveguide effective that movement of the actuator causes a light beam emitted by the light source to be directed by the lens effective to cause an overlap between the optical modes of the light beam and the optical waveguide.
  • FIG. 6 shows yet another non-limiting example of the scanning-coupling
  • a stirring mirror 608 serves to deflect the light beam from light source 602 to scan space until it overlaps the Optical mode' of optical waveguide 601 .
  • the light path 609 at one specific instant is also shown.
  • the movement of the stirring mirror may be effected by any of the means disclosed herein, including, for example, a rotating disk, a motor, a solenoid, a hydraulic mechanism, a piezoelectric mechanism, a "memory-metal" mechanism, or a manually driven actuator.
  • the light source can be controlled by turning it 'on' and Off at chosen time periods, thus controlling which of the potential pulses is actually generated while the light sources are scanning through the optical waveguides.
  • the scanning of the light source or its emitted light beam can be periodic, by constantly running the scanner in a periodic fashion, or it can be operated 'on demand,' generating anything from a single pulse to a large number of pulses.
  • the invention further comprises optical detection systems which utilize any of the scanning-coupling devices and methods described above. Additional components of such optical detection systems may include, for example, substrates for sample binding and/or processing, thermal transfer elements, thermistors, microchannels, reservoirs, electronic control boards, sample handl ing systems, an interface panel, and an enclosure or housing.
  • FIG. 8 is a block diagram of a detection system representing one possible system where the scanning-coupling systems of the current invention may be used. Particular optical scanning systems where the scanning-coupling systems of the current invention are especial ly useful are described in U.S. Patent Publ ication Nos. 2007021 1 85, published Sep. 13, 2007, and 20090068668, published Mar. 1 2, 2009.
  • the detection systems of the invention include a scanning l ight source and a substrate comprising a plurality of waveguides.
  • the scanning l ight source emits one or more light beams that are spatially translated relative to the waveguides of the substrate such that the light beam is coupled to and in optical communication with the waveguides of the substrate at some point along its scanning path.
  • spatially translated relative to the waveguides of the substrate is meant that either the scanni ng l ight source is physical ly translated through space, or that the substrate comprising the waveguides is physical ly translated through space.
  • the scanning light source is a chip referred to herein as a "scanning light source chip.”
  • the scanning light source further incl udes detector elements, it may be referred to herein as a "scanning light source/detector, ' ' or a “scanning light source/detector chip.”
  • the scanning light source chips and scanning light source/detector chips of the invention are further referred to herein as “active scanning' 1 chips in embodiments where the l ight generating elements (and detector elements when present) are integral to the chip, and as “passive scanning” chips in embodiments where the light generating elements (and detector elements when present) are external to the chip.
  • the general term “scanning light source” may be used herein to encompass any or all of these embodiments of scanning light source chips and scanning light source/detector chips. Exemplary embodiments of systems of the invention including each of these types of scanning light source are described below.
  • FIG. 7A il lustrates an exemplary detection system 700 of the invention including an active scanning light source chip 702, a substrate 704, optical sensing sites 712 and a detector 706.
  • the substrate includes excitation waveguides 708 and collection waveguides 710 that cross or intersect at an intersection region 714.
  • the active scanning light source chip 702 is at some point along its scanning path coupled to and in optical communication with one or more of the excitation waveguides 708 at a first edge of the substrate704. Additionally, the detector 706 is coupled to and in optica! communication with one or more of the collection waveguides710 at a second edge of the substrate 704. Although a single detector at one edge of the substrate is shown, it is envisioned that two or more detectors could be coupled to and in optical communication with one or more collection waveguides or excitation waveguides at various edges of the substrate (not shown).
  • a first detector could be coupled to an adjacent edge and be in optical communication with a first end of a collection waveguide, whi le a second detector could be coupled to another adjacent edge and be in optical communication with a second end of a collection waveguide.
  • a third detector can be coupled to the edge opposite to the one coupled to the active scanning light source chip and be in optical communication with the second end of the excitation waveguides (not shown).
  • the system 700 can be substantially planar.
  • the active scanning light source chip 702 can be a planar chip. This can be coupled to a planar substrate 704 that is a second chip that is further coupled to a planar detector 706 that is a third chip.
  • the system 700 is a planar lightwave circuit including three coupled chips. In one embodiment two chips are integrated into a single chip (e.g., the substrate chip and the detector chip). Such a configuration would be useful in a case where the substrate chip is reusable and can be effectively used for long periods of time. One appl ication of such a configuration would be in a system for detecting biological warfare-associated agents. In such an application it would be
  • the substrate can be a multi-element bio-analysis chip.
  • the crossing or intersecting of the excitation waveguides and the collection waveguides can be a direct physical crossing or intersecting, for example, where the excitation waveguides and the collection waveguides are embedded within the substrate in a single or in multiple layers.
  • the crossing or intersecting involves a physical space or distance between the excitation waveguides and the collection waveguides, for example, where the excitation waveguides and the collection waveguides are embedded within the substrate in separate layers.
  • the optical sensing sites 712 of the system 700 typically are associated with the intersection regions714.
  • one optical sensing site 712 is associated with each intersection region 714.
  • the number of intersection regions 714 and optical sensing sites 712 is an arrangement of 100 intersection regions 714 and 100 optical sensing sites 712. It is envisioned that the number of intersection regions and optical sensing regions on a substrate chip can be greater than 10, greater than 100, greater than 1 ,000 or greater than 10,000. It is further envisioned that the density of intersection regions can be greater than 10 per cm 2 , greater than 100 per cm 2 , greater than 1 ,000 per cm 2 or greater than 10,000 per cm 2 . In one embodiment the density of intersection regions is greater than 2,000 per cm 2 .
  • the crossing or intersecting of the excitation waveguides 708 and the collection waveguides 710 can be substantially perpendicular, for example, at an angle of 90°.
  • the crossing or intersecting can be angled less than or great than 90°.
  • a first light wave generated by the active scanning light source chip in an excitation waveguide induces the sensor to transduce an optical signal resulting in a second light wave in a collection waveguide, the second l ight wave being detectable by the detector.
  • the system 700 is a planar two-dimensional detection system.
  • the system 700 in this embodiment includes a planar active scanning l ight source chip 702 comprising a plural ity of light source elements 71 8, for example an array of switchable lasers, which is scanned perpendicular to the plane of the substrate to couple at some point along its scanning path one or more pulses of light to the plane of the substrate 704. for example, a bio- analysis chip plane.
  • the active scanning l ight source chip 702 can provide a dynamic source of light for selective and programmed excitation in respect to individual excitation waveguides 708.
  • a dynamic light source includes but is not limited to a tunable wavelength and/or tunable bandwidth l ight source. Additional ly, the system 700 of this embodiment provides for planar col lection of the emitted light from al l the excited sensing sites 712 in the collection waveguides 710, specifically in the plane of the substrate 704, such that the l ight collection is substantially perpendicular to the direction of the light produced in the excitation waveguides 708.
  • an optical sensing site 712 can be associated with each waveguide 708. It is envisioned that the number of optical sensing sites on a substrate chip can be greater than 10, greater than 100, greater than 200, greater than 1 ,000, greater than 5,000 or greater than 10,000. It is further envisioned that the density of optical sensing sites can be greater than 10 per cm 2 , greater than 100 per cm 2 , greater than 1 ,000 per cm 2 or greater than 10,000 per cm 2 . In one embodiment the density of optical sensing sites is greater than 2,000 per cm 2 .
  • a first light pulse generated by the scanning light source/detector chip in an in-coupling waveguide induces the sensor to transduce an optical signal resulting in a second light pulse in an out-coupling waveguide, the second light pulse being detectable by the detector.
  • FIG. 7B illustrates an exemplary detection system 700 of the invention including an active scanning light source/detector chip702,a substrate 704 and optical sensing sites 712.
  • the light source elements (718) can be any of a number of types of light sources including but not limited to switchable light sources or passive light sources.
  • the active scanning light source/detector chip can include in-coupling waveguides 728, out-coupling waveguides 726 and combiners 730which combine the in-coupling and the out-coupling waveguides. Such combiners 730 are well known in the art.
  • Substrate704 can include waveguides 708 and sensing sites 712 in relation to waveguides 708. For example sensing sites 712 can be on top of and in optical communication with waveguides 708.
  • the active scanning light source/detector chip can include one or more detector elements 706.
  • light source elements 71 8 are coupled to and in optical communication with in-coupling waveguides 728 on the active scanning light
  • source/detector chip spatially scans perpendicular to the plane of the substrate through a point where each of waveguides 726 is coupled to and in optical communication with its counterpart waveguide 708 on the substrate 704. At this point a l ight pulse is generated in waveguide 708 travelling from left to right. The l ight pulse interacts with the sensing area712 which acts as a transducer to create a second light pulse travell ing in waveguide 708 from right to left. This second pulse couples to the out-coupling
  • waveguides 726 on the active scanning light source/detector chip The second l ight pulse travels in waveguide 726 to the detector elements on the active scanning l ight source/detector chip.
  • FIG . 7C i l lustrates an exemplary detection system 700 of the invent ion including a passive scanning l ight source 702, connected through optical fibers 720 to an external light source comprising light source elements 7 1 8, a substrate 704. optical sensi ng sites 71 2 and a detector 706.
  • the passive light source chip 702 can include in-coupl ing waveguides or simply hold the end of the fibers.
  • Substrate 704 includes excitation waveguides 708, collection waveguides 710 and sensing sites712 on top of and in optical communication with excitation waveguides 708 and collection waveguides 71 0.
  • Detector 706 can include one or more elements 716 as described herein.
  • light source elements 71 8 are coupled to and is in optical communication with the in-coupling waveguides of the passive scanning l ight source chip 702 through a set of optical fibers 720.
  • Passive scanning light source chip 702 is at some point along its scanning path further coupled to and in optical communication with each of the excitation waveguides 708 at an edge of substrate 704.
  • detector 706 is coupled to and in optical communication with collection waveguides 710 at a second edge of substrate 704.
  • FIG. 7D illustrates an exemplary detection system 700 of the invention incl uding a passive scanning light source/detector chip 702, two sets of optical fibers 720, a light source comprising light source elements 718, a detector 706, a substrate 704, in-coupling waveguides 708 and 728, out- coupling waveguides 710 and 726 and optical sensing sites 712.
  • light source elements 71 8 are connected to and in optical communication with the in-coupling waveguides 728 on the passive scanning light source/detector chip 702 through a set of optical fibers 720.
  • detector 106 is connected to and in optical communication with the out-coupling waveguides 726 on the passive scanning light source/detector chip through a second set of optical fibers 720.
  • the passive scanning light source/detector chip at some point along its scanning path, is coupled to and in optical communication with in-coupl ing waveguides 708 on the substrate 704. At that same point along the scanning path, the out-coupling waveguides 71 0 on substrate 704 are in optical communication with the out-coupl ing waveguides 726 on the passive scanning light source/detector chip.
  • FIG. 7E illustrates a side view of an exemplary detection system 700 of the invention including a scanning light source/detector chip 702, a substrate 704 and a piezoelectric bending actuator 705.
  • the piezoelectric bending actuator is used to move the scanning light source/detector chip up and down thus scanning through a point along the scanning path where the scanning light source/detector chip is coupled to and in optical communication with the waveguides on substrate 704.
  • a first light pulse is generated in the in-coupling waveguides of substrate 704.
  • a second light pulse is coupled back from the out-coupl ing waveguides of substrate 704 to the out-coupling waveguides on the scanning l ight source/detector chip.
  • FIG . 7E illustrates the particular embodiment of the detection system shown in FIG .
  • the piezoelectric bending actuator is a representative of a variety of possible means for spatial ly translating the l ight emitted from the l ight source relative to the substrate.
  • the means for generating the relative motion of the light emitted from the l ight source relative to the substrate may incl ude a piezoelectric based motor, a step motor, an electrical motor, a magnetic actuator, a memory metal actuator, a solenoid, or a hydraulic actuator.
  • the movement of the actuator may be effected by electrical power, thermal power, magnetic power or even mechanical power (i.e., manually).
  • FIGS. 7F and 7G illustrate another exemplary chip configuration where the one or more excitation waveguides 708 exit or extend to the same edge of the chip substrate as the one or more collection waveguides 71 0.
  • the substrate has one excitation waveguide 708.
  • the substrate can have a plurality of excitation waveguides.
  • the excitation waveguides 708 can cross the collection waveguides 710 as described herein.
  • the excitation waveguides 708 can cross the collection waveguides 710 perpendicularly or approximately perpendicularly to form a plurality of optical sensing sites 7 12 at the intersections, where each optical sensing site 712 is in optical communication with both the excitation waveguide 708 and the collection waveguide 71 0.
  • the excitation waveguides 708 can bend or curve approximately 90 degrees to exit the same edge of the substrate as the collection waveguides 710.
  • the excitation waveguides 708 can be on one portion or side of the substrate while the collection waveguides 710 can be on another portion or side of the substrate.
  • the excitation waveguides 708 exit the substrate at the same side as the collection waveguides 710, only one side or edge of the substrate or chip needs to be aligned with another chip or substrate having the light sources and detectors. By reducing the number of alignments needed between the components, the manufacturing process may be simplified, thereby reducing manufacturing costs.
  • FIG. 7H illustrates another embodiment of a chip configuration that is similar to the configuration shown in FIG. 7G, but with an added loopback waveguide 713 that also exits or extends to the same edge of the chip substrate as both the excitation waveguides 708 and collection waveguides 710.
  • the loopback waveguide 71 3 can be an extension of one of the excitation waveguides 708 that loops back and extends to the edge of the chip substrate in which the other waveguides extend or exit.
  • the loopback waveguide 713 can be used to efficiently align the chip with the l ight source, detectors, optical fibers, and/or other waveguides by detecting the injected l ight with a detector at the loopback waveguide output.
  • the position of the chip can be adjusted until light is detected by the loopback waveguide detector. Without the loopback waveguide, very little light may enter the collection waveguides during alignment when no sample is present, making al ignment using the detectors more difficult.
  • FIG. 7H also illustrates an optical sensing zone 714 where the cladding has been removed to allow the light to interact with the optical sensing sites 71 2.
  • the cladding is only removed at the intersections between the collection and excitation waveguides where the optical sensing sites are located.
  • a larger segment of cladding can be removed from the excitation waveguides, starting from a point before the excitation waveguides intersect the first col lection waveguides, extending across the col lection waveguides, and ending at a point after the excitation waveguides pass the last col lection waveguides.
  • Light scattering can occur at the transition between a cladded and non-cladded portion of the excitation waveguide. This scattered l ight can enter the col lect ion waveguide and interfere with detection and measurements of light emission from the collection waveguide. By moving and eliminating the cladding transitions near the optical sensing sight, light scattering can be reduced, thereby improving the sensitivity and/or accuracy of the system.
  • the cladding transitions for the optical sensing zones 714 are located away from the optical sensing sites and are limited to a single transition before the collection waveguides and a single transition after the collection waveguides. The cladding transitions of the optical sensing zones 714 can be located far enough away from the collection waveguides to prevent, minimize or reduce light scattering interference.
  • a first scanning light source/detector chip is coupled to a first edge of the substrate and a second scanning light source/detector chip is coupled at a second edge of the substrate (not shown). It can be understood accordingly that the passage of light pulses within the devices and systems described herein, though described in terms of "left” and “right” can be practiced in a variety of directions and orientations based on the flexible arrangements of components provided herein.
  • the scanning light source chips shown in FIGS. 7A and 7C may be used in combination with the substrates shown in FIGS. 7B and 7D, together with a detector in optical communication with an opposite end of the substrate.
  • either the scanning chip or the substrate may comprise at least one combiner.
  • the scanning light source is spatially translated relative to the substrate
  • the scanning of the emitted l ight relative to the waveguides of the substrate may also be effected by spatially translating the substrate relative to the light source, or by spatially translating any part or component of the scanning l ight source, such as one or more mirrors, lenses, or prisms, using any of the means disclosed herei n.
  • optical sensing site 712 can be associated with each optical sensing site 712 .
  • the number of optical sensing sites on a substrate chip can be greater than 10, greater than 100, greater than 200, greater than 1 ,000, great than 5,000 or greater than 10,000. It is further envisioned that the density of optical sensing sites can be greater than 10 per cm", greater than 1 00 per cm 2 , greater than 1 ,000 per cm 2 or greater than 10,000 per cm 2 . In one embodi ment the density of optical sensing sites is greater than 2,000 per cm 2 .
  • a first light pulse generated by the scanning light source chip in an in-coupling or excitation waveguide induces the sensor to transduce an optical signal resulting in a second l ight wave in an out-coupling or col lection waveguide, the second light wave being detectable by the detector.
  • FIG. 8 is an exemplary illustration of the detection system of the invention as part of a working system 801 in a housing 809. Whi le the detection system i l l ustrated in FIG . 7A-7E is the core of the present invention, in order to facilitate the operation of this system, one or more other modules can be included in a working system that includes the detection system components of the invention.
  • FIG. 8 i l lustrates one possible configuration for a working system 801 that can include housing 809 for enclosing various modules of working system 801 including but not limited to substrate 804, robotic system 803, scanning light source chip802, multi-element detector 806, electronic boards 807 and interface panel 805. Substrate 804, scanning l ight source chip802, and multi-element detector 806, are discussed in detail below.
  • housing 809 in one embodiment an enclosure or housing 809 holds in place two fixed chips (e.g., of a 3-chip architecture), namely, scanning light source chip 802 and multi-element detector 806. Accordingly, in this embodiment substrate chip 804 is movable in relation to scanning light source chip 802 and multi-element detector 806. Housing 809 can include any number of accurately machined parts and or components as described herein, allowing, for example, the relative alignment of the 3 optical chips, and the movement of the scanning light source chip perpendicular to the plane of the substrate.
  • the working system housing can optionally include temperature control and vibration isolation for the working system (not shown).
  • working system 801 can further include an X, Y, Z, ⁇ robotic system 803 for positioning substrate 804 as required within working system 801 .
  • X, Y, Z, ⁇ robotic system 803 can be a translation stage with several degrees of freedom for receiving or accepting substrate 804, holding it in place, and aligning it in relation to the rest of working system 801 .
  • ⁇ robotic system 803 can eject substrate 804 from working system 801 .
  • the working system can further include an al igning system (not shown).
  • An aligning system can include one or more light sources, one or more detectors and one or more cameras for active detection of the position of the substrate of the invention. Based on the detected position, the aligning system can align the substrate to the rest of the working system modules, for example, to provide aligned optical communication between the substrate and the scanning light source/detector chip.
  • working system 801 can further incl ude one or more electronic boards 807, for example, an electronic driving board and a control board. It is envisioned that one or more electronic boards can control all the different parts of the working system. Electronic boards 807 can control scanning light source 802 and any other light source present in the system. Electronic boards 807 can be adapted to read any or all of the detectors and cameras in working system 801 .
  • Electronic boards 807 can further be adapted to drive robotic system 803 and control its motion, to control the motion of the scanning light source chip, and optionally to monitor and control temperature in different areas of the system.
  • Electronic boards can incl ude logic elements and processors (not shown). It is envisioned that electronic boards can further include embedded software both for control ling the working system and for interfacing the outside world, for example by way of interface panel 805 which can include a key-pad or any other input/output port.
  • working system 801 can additional ly incl ude one or more interface panel 805. It is anticipated that the system wi ll have one or more interface panel 805 to allow a user to interface with the system and operate it.
  • Interface panels can include any number of input and output ports well known in the art for connecting the system to other systems or to an external control console (not shown).
  • FIG. 9A illustrates an exemplary substrate 904 of the second embodiment of the detection system of the current invention (as show in FIG. 7B) further including barriers 91 1 intended to block stray light within the substrate and reduce crosstalk between the different elements of the substrate.
  • Barriers 91 1 can be light absorbing or light reflecting.
  • Barriers 91 1 can be variously sized, shaped and positioned between waveguides 908 in any of a number of orientations to achieve a desired optical effect. As shown in FIG. 9A, barriers 91 1 can be arranged between two adjacent waveguides and proximal to optical sensing sites912.
  • Waveguides 908 are used to guide a primary l ight wave (excitation light; see dashed arrow) from the left edge of substrate 904 to optical sensing sites 912. Waveguides 908 then guide a secondary light wave (collected at optical sensing sites 912; see dashed arrow) from optical sensing sites 912 back to the left edge of substrate 904.
  • a primary l ight wave excitation light; see dashed arrow
  • Waveguides 908 then guide a secondary light wave (collected at optical sensing sites 912; see dashed arrow) from optical sensing sites 912 back to the left edge of substrate 904.
  • FIG. 9B illustrates an exemplary substrate 904 of the fourth embodiment of the detection system of the current invention (as shown in FIG. 7D) further including in-coupl ing waveguides 908 and combiners 926.
  • the primary light wave (excitation light; see dashed arrow) is coupled to
  • Barriers 1 1 have the same purpose as described above in FIG. 9A.
  • FIG. 9C schematical ly il l ustrates a cross section of the substrate 904 of one embodiment of the current invention.
  • in/out coupling waveguides 908 are embedded underneath a surface of substrate 904.
  • Optical sensing sites 912 can be etched into a surface, for example, the upper cladding of substrate 904 and located, for example, ad jacent to and on top of
  • waveguide 908 facil itating optical communication between them.
  • the optical sensing sites can also be located on the surface of substrate 904 or etched only part of the way into the upper cladding, or all the way through the waveguides (not shown).
  • the waveguides can be single-mode waveguides, multi-mode waveguides or any combination of the two, namely, single mode in the vertical dimension and multi-mode in the lateral dimension.
  • FIG. 9D i llustrates an exemplary substrate 904 of the first and third embodiments of the detection systems of the current invention (as shown in FIGS. 7A and 7C), further including
  • barriers 91 1 intended to block stray l ight withi n the substrate and reduce crosstal k between the different elements of the substrate.
  • the barriers 91 1 can be l ight absorbi ng or l ight reflecting.
  • the barriers 91 1 can be variously sized, shaped and positioned between the col lection waveguides 91 0 and/or the excitation waveguides 908 in any of a number of orientations to achieve a desired optical effect. As shown in FIG. 9D, the barriers 91 l ean be arranged in a row between two adjacent col lection waveguides and proximal to the optical sensing sites 912 and intersection region 914.
  • the substrate 904 can include excitation waveguides 908 and col lection waveguides 910 embedded beneath a surface of the substrate 904 in multiple layers. As shown, the excitation waveguides 908 cross, physically intersect, and are in optical communication with the collection waveguides 910 at the intersection regions 914. In the embodiment shown in FIG . 9E, the optical sensing site 912is positioned at the intersection region 914 above and in optical communication with the excitation waveguides 908. As further shown in FIG.
  • the substrate 904 includes multiple layers including a silicon layer 920 and a silica (Si02) layer 922, wherein the excitation waveguides 908 and the collection waveguides 910 are embedded within the silica (Si02) layer 922.
  • the substrate can include excitation waveguides 908 and collection waveguides910 embedded underneath a surface of the substrate 904 in a single layer. As shown, the excitation waveguides 908 cross, physically intersect and are in optical communication with the collection waveguides 910. In contrast to the embodiment shown in FIG. 9E, here the intersection between excitation waveguides 908 and collection waveguides 910 occurs internal to the collection waveguides 91 0. As further shown in FIG. 9F, the substrate 904 includes multiple layers including a sil icon layer 920, a silica (Si02) layer 922, and a cladding layer 924.
  • the excitation waveguides 908 and collection waveguides 910 can be embedded within the silica (Si02) layer 922.
  • the optical sensing site 912 can be embedded within both the cladding layer 924 and the silica (Si02) layer 922.
  • the optical sensing site can be embedded solely within the cladding layer (not shown).
  • the excitation waveguides and collection wave guides can be single- mode or multi-mode waveguides.
  • the excitation waveguides are single-mode and the collection waveguides are multi-mode.
  • waveguide configurations can include single- or multi-mode configurations in either vertical or lateral orientations within a waveguide.
  • the excitation waveguides 908 can support a single mode in the vertical dimension and multi modes in the lateral dimension.
  • the excitation waveguides 908 and the col lection waveguides 910 can span the entire substrate from one edge to another edge.
  • the substrate 904 components and optical sensing sites 912 can include dimensions.
  • FIG. 9F shows two cross-section views of the substrate 904.
  • View AA is a cross- section view in plane A as indicated in FIG. 9D and FIG. 9E.
  • View BB is a cross-section view in plane B as i ndicated in FIG. 9D and FIG . 9E.
  • the thickness of the cladding layer 924 above the excitation waveguides can be about 0. 1 ⁇ to about 20 ⁇ . In one embodiment the cladding layer 924 thickness is about 1 ⁇ to about 2 ⁇ .
  • an opening of the optical sensing site 912 can include the following dimensions: about 20 ⁇ by about 2 ⁇ .
  • the distance between collection waveguides 910 can range from about 1 ⁇ to about 1000 ⁇ .
  • the distance between collection waveguides 910 can be about 100 ⁇ .
  • the distance between collection waveguides 910 and the silicon layer 920 can be about 1 ⁇ to about 1 00 ⁇ .
  • the distance between collection waveguides 910 and the silicon layer 920 can be about 10 ⁇ to about 20 ⁇ .
  • the excitation waveguides 908 and collection waveguides 910 can be channel waveguides. Exemplary ranges for waveguide dimensions in the embodiment shown in FIGS. 9E and 9F include about 0. 1 to about 1 0 ⁇ thick and about 1 to about 100 ⁇ wide.
  • the excitation waveguides 908 can include cross-section dimensions of about 0.1 ⁇ by about 100 ⁇ and the collection waveguides 910 can include cross- section dimensions of about 0.2 ⁇ by about 1 00 ⁇ .
  • FIG. 9G in a side view illustrates another embodiment of substrate 904 of the invention in relation to a thermal transfer element 903, for example, a thermoelectric cooler (TEC).
  • Thermal transfer element 903 is a temperature control system useful for heating or cooling a chip, for example, substrate 904.
  • the thermal transfer element may be referred to herein as a cooling element, it is to be understood that where the thermal transfer element is configured to increase and decrease the temperature of a chip, the component functions essentially as a heating and as a cooling element depending on the induced direction of the electrical current.
  • the thermal transfer element can provide a range of useful temperatures.
  • the thermal transfer element can be configured to provide a temperature in the range between about -40° C. to about 120° C. as desired.
  • the thermal transfer element can be configured to provide a temperature in the range between about -40° C. to about 120° C. as desired.
  • the transfer 903 element can be adapted to receive substrate 904 of the invention.
  • the thermal transfer element 903 can be adapted to contact part or al l of a surface of the substrate 904 of the invention.
  • thermal transfer element 903 in conj unction with substrate 904 of the invention is useful, for example, for the amplification of tested sample molecules through processes such as the polymerase chain reaction (PCR) as described herein.
  • PCR polymerase chain reaction
  • the embodiment as described for FIG. 9G provides the capability of controll ing the temperature of the entire substrate such that as the temperature of the entire substrate is cycled, samples at any optical sensing site can be amplified by PCR simultaneously.
  • FIG. 9H illustrates another embodiment of substrate 904 of the invention wherein optical sensi ng site 91 2 includes heater 905and thermistor 907.
  • optical sensing site 912 of substrate 904 can include heater 905, for example, a thin-film heater, in the vicinity of each sensing sites 912.
  • Heater 905 can be adapted to enable individual temperature control for each sensing site 912.
  • thermistor 907 can be located at or near each sensing site 91 2 thereby providing for measuring the local temperature.
  • this embodiment provides the capability of running the same or any desired different number of cycles and the same or any desired different temperature profiles for each and every sensing site.
  • optical sensing site 912 of substrate 904 can include heater 905, for example, a thin-fi lm heater, in the vicinity of one or more sensing site 912.
  • Heater 905 can be adapted to enable individual temperature control for each sensing site 912.
  • thermistor 907 can be located at or near one or more sensing site9 ! 2 thereby providing for measuring the local temperature.
  • this embodiment provides the capability of running the same or any desired different number of cycles and the same or any desired different temperature profiles for each and every sensing site.
  • FIGS. 9G, 9H, and 91 can support real- time PCR.
  • signal detection these both embodiments (see FIGS. 9G, 9H, and 91) can be done while the samples are in the process of the amplification cycles, thereby enabling real time analysis of the PCR process.
  • FIG. 9J illustrates yet another embodiment of substrate 904 of the invention wherein substrate 904 additionally includes reservoirs 913 and microchannels 909 in relation to optical sensing sites 912.
  • microfluidics are incorporated into the substrate.
  • Microfluidics can be adapted to drive liquid (in this case the tested sample) using the capillary effect across the substrate. As illustrated in FIG. 9J, this can be achieved by an arrangement of microchannels 909, optionally of varying width, which force the sample from one or more reservoirs 913 to optical sensing sites 912 which can include etched wells to receive the sample.
  • the microchannels can be either etched on the face of the chip itself or can be added as an external structure on a surface of the substrate.
  • FIG. 9K illustrates yet another embodiment of substrate 904 of the invention wherein substrate 904 additionally includes reservoirs 91 3 and microchannels 909 in relation to optical sensing sites 912.
  • microfl uidics are incorporated into the substrate.
  • Microfluidics can be adapted to drive liquid (in this case the tested sample) using the capi llary effect across the substrate.
  • this can be achieved by an arrangement of microchannels 909, optionally of varying width, which force the sample from one or more reservoirs 913 to optical sensing sites 912 which can include etched wells to receive the sample.
  • the microchannels can be either etched on the face of the chip itself or can be added as an external structure on a surface of the substrate 904.
  • a sample to be tested can be pipetted into a reservoir at one end of the substrate.
  • the sample can then be distributed using the microfluidic system to the optical sensing sites and sensing wells where it is al lowed to bind to pre-spotted probes and can subsequently be optically detected and analyzed.
  • Several reservoirs may be used to separate different samples/patients or for running several parallel tests.
  • the substrate of the system may be dip-coated with one or more probes configured to interact biochemical ly with a desired biological ly active analyte molecule.
  • Example 1 describes chip coating protocols for antibody or ol igonucleotide attachment.
  • one or more probes may be appl ied to a sensor of the optical sensi ng sites using a print head.
  • delivery of sample to the optical sensing sites of the system comprises del ivering the sample using an assay head.
  • One possible print head technology is described in U.S. patent application Ser. No. 1 1 /241 ,060, filed on Sep. 30, 2005, and U.S. patent application Ser. No. 1 1 /632,086, filed on Jul . 6, 2005.
  • FIG. 10A in a top view illustrates an exemplary substrate 1004 of the system of the invention wherein the collection waveguides 1010 include funnels 1017 (shown in detai l in FIG. 10B) for collecting light.
  • the substrate 1004 can include a 10x 10 array consisting of 10 excitation waveguides 1008 (e.g., about 5 ⁇ wide by about 2 ⁇ deep), 10 collection waveguides 1010 (e.g., about 30 ⁇ wide by about 1 0 ⁇ deep), 100 optical sensing sites 1012 (e.g., wel ls about 30 ⁇ long by about 5 ⁇ wide by about 10 ⁇ deep), 100 funnelsl 017 for collecting light from the optical sensing sites 1012 and barriers 101 1 (e.g., light absorbing channels) to reduce crosstalk between the optical sensing sites 1012.
  • the substrate can include greater than 10, greater than 100 or greater than 1 ,000 excitation waveguides 1008 and collection waveguides 1010.
  • excitation light can be coupled into one or more excitation waveguides 1008 on the left hand side of the substrate 1004 from, for example, a scanning light source chip.
  • Excitation l ight can travel along the excitation waveguides 1008 and couple into the optical sensing sites (e.g., wells) through an evanescent field tail.
  • Excited fluorescence generated in the optical sensing site 1012 can be col lected along the long facet of the optical sensing site 1012into the funnels 1017.
  • the funnels 1 017 can channel the light into the collection waveguides 1010.
  • the light in the collection waveguides 101 0 can be coupled out at the "bottom" of the substrate 1004 into a detector array (not shown).
  • Light scattered outside the optical sensing sites 1012 can be blocked by a series of barriers 1 01 1 (e.g., light absorbers) to avoid crosstalk between parallel collection waveguides 1010.
  • the substrate shown in FIG. 10A includes two waveguide layers.
  • a bottom layer about 2 ⁇ thick, can include the excitation waveguide 1008.
  • the bottom layer can have a higher refractive index in order to increase the evanescent field tail presence in the optical sensing sites.
  • An upper layer about 10 ⁇ thick, can contain the optical sensing site and the light collection structures (funnels and waveguides).
  • the upper layer can have a lower refractive index than the bottom layer in order to minimize l ight loss when coupling the light out of the substrate to the detector.
  • both the excitation and collection waveguides are multimode.
  • the excitation waveguides 1 008 can be thinner than the collection waveguides 1 010.
  • the excitation waveguides 1008 can have a width of about 5 ⁇ (see FIG. 1 0B) and a height of about 2 ⁇ (see FIG. I OC).
  • the col lection waveguides 1010 can have a width of about 30 ⁇ (see FIGS. 10A and 10B) and a height of about 1 0 ⁇ (see FIG. I OC).
  • the optical sensing sites can be wells that are narrow (about 5 ⁇ ) and long (about 30 ⁇ ) with light collectable along the long facet. Such a configuration increases the efficiency of light collection.
  • light excitation coupling into the well can increase due to the long coupling length.
  • the wel l dimensions (5*30x 10 ⁇ 3 ) yield a volume of 1 .5 pico-liter. Larger wells are also envisioned in a variety of sizes yielding volumes ranging from about 0.1 pico-liter to about 100 micro-liter.
  • the funnel can have a radius for the collection, confinement and coupling of light into the collection waveguides. Suitable ranges for radii can include from about 100 ⁇ to about 1000 ⁇ ⁇ .
  • the barriers 101 1 as illustrated in FIGS. 10A and 10B can be trenches filled with light absorbing material (e.g., a metal such as gold). Where the barriers 101 1 are trenches, the trenches can include openings above the excitation waveguide 1008 to avoid loss at the crossing points (not shown).
  • light absorbing material e.g., a metal such as gold
  • the overall dimensions of the substrate illustrated in FIG. 10A can be about 1.2 by about 1 .2 mm 2 . Margins can optionally be included around the substrate to adjust the overall dimensions as desired.
  • FIG. 1 1 A i llustrates an exemplary substrate 1 104 of the system of the invention wherein the excitation waveguides 1 ! 08include a plurality of branches 1 121 (shown in detail in FIG. 1 I B) for tapping light from the excitation waveguides and coupling it into the sensing wells.
  • the substrate 1 104 can be made up of several waveguide layers (e.g.. three waveguide layers). Such a configuration can be useful, for example, to optimize excitation and fl uorescence collection while minimizing loss and crosstalk.
  • FIGS. 1 1 C and 1 1 D are schematic cross-section views of the substrate 1 104 through planes at (AA) and (BB) respectively as indicated in FIG. 1 1 B.
  • the substrate consists of three waveguide layers having core refractive index of 1 .7 and clad reflective index of 1 .4.
  • Useful core refractive index values range from about 1 .45 to about 2.
  • useful clad refractive i ndex val ues range from about 1 .4 to about 1 .5.
  • a first bottom layer can be about 10 ⁇ thick and include the collection waveguides 1 1 10.
  • the collection waveguides 1 1 10 can be about 30 ⁇ wide, multimode and traverse the substrate 1 104 from substantially edge to edge.
  • a second middle waveguide layer can be about 0.5 ⁇ to about 1 ⁇ thick and include coupl ing waveguide branches 1 1 21 (see FIGS. 1 0A and 10B).
  • the branches 1 121 can couple excited light into the optical sensing sites, which can be wel ls.
  • FIG. 14A il lustrates an active scanning light source chip 1402 of the first embodiment (see FIG. 7A) of the detection system of the current invention.
  • the active scanning light source chip 1402 i ncludes l ight source elements 141 8 and in-coupling waveguides 1440.
  • a primary light wave generated by the light source elements is coupled from left into in-coupling waveguides 1440.
  • Waveguides 1440 guide the primary light wave to the right edge of the active scanning light source chipl 402 and couple it out to the substrate (not shown).
  • FIG. 14B i llustrates an active scanning light source/detector chip 1402 of the second embodiment (see FIG. 7B) of the detection system of the current invention.
  • the active scanning light source/detector chip 1402 includes light source elements l 418, detector elements 1416, in-coupling waveguides 1440, out-coupling waveguides 1438 and combiner 1436.
  • a primary light wave (excitation light) generated by the light source 1418 is coupled from left into in-coupling waveguides 1440.
  • the excitation light is then combined by combiners 1436 into out-coupling waveguides 1438 which then guide it to the right edge of active scanning light source/detector chip 1402 and couple it out to the substrate (not shown).
  • the substrate couples back the secondary light wave (collected at the optical sensing sites— not shown) to out-coupling waveguides 1438at the right edge of active scanning light source/detector chip 1402.
  • the secondary light wave is guided by the out-coupling
  • FIG. 12A illustrates a passive scanning light source chip 1202 of the third embodiment (see FIG. 7C) of the detection system of the current invention.
  • Passive scanning light source
  • chip 1202 includes in-coupling waveguides 1228.
  • a primary light wave (excitation light) is coupled from left into in-coupling waveguides 1228 through a set of optical fibers (not shown).
  • Waveguide l 228 guides the primary light wave to the right edge of the passive scanning light source chip 1202 and couples it out to the substrate (not shown).
  • FIG. 12B il lustrates a passive scanning light source/detector chip 1202 of the fourth embodiment (see FIG. 7D) of the detection system of the current invention.
  • the passive scanning light source/detector chip 1202 includes in-coupling waveguides 1228, out-coupling waveguides 1226 and combiners 1230.
  • a primary l ight wave (excitation light) is coupled from the left into in-coupling waveguides 1228 through a set of optical fibers (not shown).
  • the excitation light is then combined by combiners 1230 into the out-coupling waveguides 1226 which then guide it to the right edge of passive scanning l ight source/detector chip 1 202 and couple it out to the substrate (not shown).
  • the substrate couples back the secondary light wave (col lected at the optical sensing sites— not shown) to out- coupling waveguides 1226 at the right edge of passive scanning light source/detector chip 1202.
  • the secondary l ight wave is guided by waveguides 1 226 from right to left and is coupled out of passive scanning l ight source/detector chip 1202 at its left edge to the detector (not shown) through a set of optical fi bers (not shown).
  • FIG. 12C illustrates a second possible design of a passive scanning light source/detector chip 1202 of the fourth embod iment (see FIG. 7D) of the detection system of the current invention.
  • the passive scanning l ight source/detector chip 1202 includes in-coupl ing waveguides 1 228 and out-coupling waveguides 1226.
  • a primary l ight wave (excitation l ight) is coupled from left into in-coupling waveguides 1228 through a set of optical fibers (not shown). The excitation light is then guided to the right edge of passive scanning light source/detector chip 1202 where it is coupled out to the substrate (not shown).
  • the substrate couples back the secondary light wave (collected at the optical sensing sites— not shown) to out-coupl ing waveguides 1226 at the right edge of passive scanning light source/detector chip 1202.
  • the secondary light wave is guided by waveguides 1226 from right to left and is coupled out of passive scanning l ight source/detector chip 1202 at its left edge to the detector (not shown) through a set of optical fibers (not shown).
  • the light source elements 718 in FIGS. 7A- 1 D can comprise a dynamic light source allowing for selective and programmed generation of the primary light wave in one or more individual elements.
  • the light source elements 718 can provide variable wavelengths of light.
  • the light source element is a broad-band source. In another embodiment, the light source element is a tunable source.
  • the number of light source elements 718 will be equal to the number of excitation waveguides in the substrate of the system.
  • the interface between the scanning light source outputs should match, in terms of pitch, the excitation waveguides in the substrate to allow these two elements at some point along the scanning path to efficiently couple and transfer light from the scanning light source chip to the excitation waveguides or to the in-coupling waveguides of the substrate.
  • the light source elements depicted in FIGS. 7A-D and FIGS. 14A and 14B can include different types of light generating elements.
  • the light generator elements are light emitti ng diodes (LEDs).
  • the light generator elements are laser diodes (LDs).
  • Each individual l ight generator element is separately controlled and can be turned on or off as desired.
  • the scanning l ight source chip includes 10 or more light generator elements.
  • the scanning l ight source chip includes 1 00 or more light generator elements.
  • the scanning light source chip includes 1000 or more light generator elements.
  • the scanning light source chip includes between 10 and 100 light generator elements.
  • the detector elements depicted in FIGS. 7A-D and FIGS. 14A and 14B can include different types of detector elements.
  • the detector elements are PIN diodes.
  • the detector elements are avalanche photo-diodes (APD).
  • the detector elements are a group of pixels which are part of a CCD array. Each individual detector element is separately controlled and read.
  • the scanning light source chip includes 10 or more detector elements.
  • the scanning light source chip includes 100 or more detector elements.
  • the scanning l ight source chip includes between 1 0 and 100 detector elements.
  • the detector element has a spectral range of between 400 to 1 000 nm, a photosensitivity (A/W) of >0.3, an active area per element of 0.005 mm 2 , 128 elements, and a pitch of ⁇ 0.1 mm.
  • the detector is a silicon photodiode (PN, PIN, CCD or APD) array.
  • PN silicon photodiode
  • An example of a suitable detector array is the Hamamatsu 64x2048 CCD chip (PN— S I 0420-1 106).
  • the light source elements and detector elements on the scanning l ight source chip can be integrated on a single chip which includes an array of two or more light source elements, an array of two or more detector elements, an array of two or more in-coupling waveguides, an array of two or more out-coupling waveguides and an array of two or more combiners.
  • each light source element is optically coupled to one in-coupling waveguide and adapted such that most of the l ight emitted by the light generator element propagates along that waveguide.
  • the waveguides can extend to the edge of the chip where they can be brought at some point along the scanning path of the chip to couple the light propagating within them to the substrate.
  • two light source elements each optionally emitting at a different wavelength can be coupled to a single in-coupling waveguide.
  • more than two light source elements each optionally emitting at a different wavelength can be coupled to a single in-coupling waveguide.
  • the light source elements on the scanning light source chip can be integrated on a single chip which includes an array of two or more light source elements and an array of two or more waveguides.
  • each light source elements is optically coupled to one waveguide and adapted such that most of the light emitted by the light source element propagates along the waveguide.
  • the waveguides can extend to the edge of the chip where they can be brought to couple the light propagating within them to the substrate.
  • two light source elements each optionally emitting at a different wavelength can be coupled to a single waveguide.
  • more than two l ight source elements, each optionally emitting at a different wavelength can be coupled to a single waveguide.
  • the scanning l ight source chip can include, in addition to light source elements, detector elements and waveguides, light manipulating features such as lenses, filters, switches, modulators, spl itters, combiners, mirrors and circulators.
  • the control of the scanning light source chip can be either integrated on the same chip as the l ight source elements, detector elements and waveguides or alternatively can be external to the chip.
  • the scanning l ight source chip can have an electrical interface to an external driver or external controller or logic interface to an external control system.
  • the control of the light source elements and detector elements al lows driving each l ight source element and each detector element separately. It further al lows also control of the other features present on the scanning l ight source chi p such as, for example, the modulators and switches.
  • Additional elements useful in planar l ightwave circuits incl uding but not limited to couplers, fi lters, mirrors, circulators, splitters, modulators, switches and trenches are envisioned as part of the system described herein (not shown).
  • Such elements when integrated into the substrate or into the scanning light source chip can serve to manipulate the incoming first light waves in the in-coupl ing waveguides or outgoing second light waves in the out-coupling waveguides.
  • such elements when integrated into the substrate or into the scanning l ight source chip can serve to manipulate both incoming first l ight waves in the excitation waveguides or outgoing second light waves in the collection waveguides.
  • a range of dimensions for the various features described herein include: waveguides thickness— about 20 mil to about 50 ⁇ ; waveguide width— about 1 ⁇ to about 500 ⁇ ; waveguide length— about 1 mm to about 100 mm; optical sensing site length— about 100 ⁇ to about 100 mm; optical sensing site width— about 1 ⁇ to about 500 ⁇ ; optical sensing site depth— about 0 ⁇ to about 20 ⁇ ; waveguide pitch— about 10 ⁇ to about 10 mm; substrate thickness— about 1 00 ⁇ to about 5 mm; upper cladding thickness— about 0 ⁇ to about 20 ⁇ ; and lower cladding thickness— about 0.1 ⁇ to about 20 ⁇ .
  • the substrate of the detection system can made up of any of a number of well known materials suitable for use in planar lightwave circuits.
  • useful substrate materials include but are not limited to silica (Si02), glass, epoxy, lithium niobate and indium phosphide as wel l as combinations thereof.
  • the waveguides disclosed herein can be made up of silicon, silica (Si02) and derivatives thereof, si licon oxynitride (SiON) and derivatives thereof, silicon nitride (SiN) and derivatives thereof, tantalum oxide (TaO x ) and its derivatives thereof, polymers, lithium niobate and indium phosphide as well as combinations thereof.
  • UV light is used to change the refractive index of a waveguide material after deposition.
  • the silicon layer 1326 can be made up of a silicon wafer having a thickness from about 0.1 mm to about 1 0 mm.
  • the silicon wafer can have a thickness from about 0.3 to about 1 mm.
  • the silicon wafer has a thickness of 0.65 mm.
  • the silica (Si02) layer 1322 is a 14 ⁇ thermal oxide layer of silica (Si02) created by placing the silicon in an oxygen-rich environment inside a furnace at high temperature.
  • the top sil icon layer oxidizes over time (several hours) creating a Si02 layer.
  • the cladding layer 1324 is 15 ⁇ thick and deposited by a PECVD (Plasma-Enhanced Chemical Vapor Deposition) process after etching to produce the waveguides 1308.
  • the various layers of the substrate can include different refraction index properties.
  • a waveguide layer e.g. SiN
  • a cladding layer of si l ica deposited thereon has a higher refraction index than a cladding layer of si l ica deposited thereon.
  • the substrate 1304 can include two waveguides 1 308 arranged for l ight wave coupling on a si lica (Si02) layer 1322.
  • the substrate 1304 can include two waveguides 1 308 arranged for l ight wave coupling on a si lica (Si02) layer 1322.
  • the waveguides 1 308 can be arranged for guiding uncoupled light waves on a si lica (Si02) layer 1 322and over-clad with a cladding layer 1 324.
  • the optical sensing sites in one embodiment are in the form of wells, for example, etched wel ls (see FIG. 9C cross-section view). Where the optical sensing site is a well, it can act as a vessel for a l iquid sample. In another embodiment the optical sensing sites are a region on the surface of the substrate, for example, above the waveguides. In a further embodiment, the optical sensing sites are biochemical interaction sites.
  • the optical sensing site is a well containing a sensor single stranded DNA ol igonucleotide having a fluorescent tag attached
  • a solution containing a target complementary single stranded DNA added to the well could biochemically interact by base-pairing with the sensor within the optical sensing site (not shown).
  • the optical sensing site is a location or well containing one or more immunoassay reagent for conducting an immunoassay as described herein.
  • the optical sensing sites comprise optical transducers (not shown).
  • An optical transducer is defined as any device that generates a measurable change (wavelength, amplitude or phase) to the incoming primary light wave which can thus be monitored in the outgoing secondary light wave.
  • the optical transducers are fluorescence wells including fluorescent or luminescent compounds, wherein light waves guided by the waveguides excite the fluorescent or l uminescent compound in the wells in the presence of a target, and the same waveguides collect and guide light emitted from the wells to the detector (possibly through an adapter chip), for example at the edge of the chip (not shown).
  • the sensor of the optical sensing site of the system can be a sensor that discriminates or interacts with a target (e.g., a biologically active analyte) in a sample from, for example, a biological, man-made or environmental source.
  • a first lightwave can induce the sensor to transduce an optical signal to a second light wave.
  • a measurable change in the second light wave can result when the sensor discriminates or interacts with the target.
  • the presence of the target in the sample is indicated.
  • any of a number of sensors can be used with the detection system to measure phenomena associated with the sensing of a target in a sample.
  • suitable sensors include, but are not l imited to, a fluorescence well or cell, an absorption cell, an interferometric sensor, a diffractive sensor or a surface plasmon resonance (SPR) detector.
  • the measurable phenomenon can be light emission from luminescent or fluorescent molecular tags. For example, emitted light at an altered wavelength can be measured.
  • changes in the sample optical density (OD) can measurably affect the intensity of the light passing through the sample.
  • changes in the effective refractive index of a waveguide generate a phase difference between two light waves leading to different interference patterns measurable as a difference in i ntensity at the detector.
  • changes in the effective refractive index at the surface of a diffract ive element affect the diffraction angle of the light for a given wavelength or alternatively affect the wavelength at a given diffraction angle.
  • changes in the effect ive refractive index at a metal-dielectric interface affect the resonance conditions for generating surface plasmons.
  • the control system can manage steps such as aligning and driving the scanning l ight source chip to scan the edge of the substrate, in addition to switching the light output from the light source, reading the detector array and reporting the results detected.
  • Biologically active analyte molecules in this context include any of the biologically active analyte molecules disclosed herein.
  • Sample preparation suitable for use with the system and methods described herein can include any of a number of well known methods for collection and analysis of biological and/or environmental samples.
  • the sample can be, for example, manipulated, treated, or extracted to any desired level of purity for a target of interest.
  • the sample can be a bodily fluid suspected of containing a biologically active analyte.
  • bodily fluids include but are not l imited to blood, serum, saliva, urine, gastric and digestive fluid, tears, stool, semen, vaginal fluid, interstitial fluids derived from tumorous tissue, and cerebrospinal fluid.
  • the systems described herein can be used for screening a large variety of samples.
  • the sample may originate from body fluids as discussed. Methods of obtaining samples include but are not limited to cheek swabbing, nose swabbing, rectal swabbing, skin fat extraction or other collection strategies for obtaining a biological or chemical substance.
  • the sample may originate from any substance in a solid phase, liquid phase or gaseous phase.
  • the sample may be col lected and placed onto the substrate or the substrate may be directly exposed to the investigated sample source (e.g. water reservoir, free air) and interact with it.
  • the bodily fluids are used directly for detecting one or more biologically active analyte present therein without further processi ng.
  • the bodily fl uids can be pre-treated before performing the analysis with the detection system.
  • the choice of pre- Ireat ments wi l l depend on the type of bodily fluid used and/or the nature of the biologically active analyte under investigation. For instance, where the biological ly active analyte is present at a low level in a sample of bodily fluid, the sample can be concentrated via any conventional means to enrich the biologically active analyte.
  • Methods of concentrating a biologically active analyte include but are not l imited to drying, evaporation, centrifugation, sedimentation, precipitation, and amplification.
  • the biologically active analyte is a nucleic acid
  • it can be extracted using various lytic enzymes or chemical sol utions according to the procedures set forth in Sambrook et al . ("Molecular Cloning: A Laboratory Manual"), or using nucleic acid binding resins following the accompanying instructions provided by manufactures.
  • extraction can be performed using lysing agents including but not limited to denaturing detergents such as SDS or nondenaturing detergents such as thesit (2-dodecoxyethanol), sodium deoxylate, Triton® X- 100, and Tween® 20.
  • denaturing detergents such as SDS or nondenaturing detergents such as thesit (2-dodecoxyethanol), sodium deoxylate, Triton® X- 100, and Tween® 20.
  • pretreatment can include diluting and/or mixing the sample, and filtering the sample to remove, e.g., red blood cells from a blood sample.
  • Targets detectable using the detection system include but are not limited to, a biologically active analyte including a nucleic acid, a protein, an antigen, an antibody, a microorganism, a gas, a chemical agent and a pollutant.
  • a biologically active analyte including a nucleic acid, a protein, an antigen, an antibody, a microorganism, a gas, a chemical agent and a pollutant.
  • the target is a nucleic acid that is DNA, for example, cDNA.
  • the DNA target is produced via an amplification reaction, for example, by polymerase chain reaction (PCR).
  • the detected biologically active analyte is a protein representing a known biomarker for a disease or specific condition of the investigated organism.
  • several different biologically active analytes can be proteins provided as a panel of bio-markers wherein relative concentrations of the bio-markers are indicative for a disease or other condition of the investigated organism.
  • the target is a microorganism that is a pathogen.
  • the target is a chemical agent, for example, a toxic chemical agent.
  • the target is a nucleic acid
  • it can be single-stranded, double-stranded, or higher order, and can be linear or circular.
  • exemplary single-stranded target nucleic acids include mRNA,
  • target nucleic acids include genomic DNA, mitochondrial DNA, chloroplast DNA, dsRNA or dsDNA viral genomes, plasmids, phage, and viroids.
  • the target nucleic acid can be prepared synthetically or purified from a biological source. The target nucleic acid may be purified to remove or diminish one or more undesired components of the sample or to concentrate the target nucleic acids. Conversely, where the target nucleic acid is too concentrated for the particular assay, the target nucleic acid may be di luted.
  • the nucleic acid portion of the sample comprising the target nucleic acid can be subjected to one or more preparative reactions.
  • These preparative reactions can include i n vitro transcription ( I VT), labeling, fragmentation, ampl ification and other reactions.
  • mRNA can first be treated with reverse transcriptase and a primer to create cDNA prior to detection and/or ampl i fication; this can be done in vitro with puri fied mRNA or in situ, e.g. in cells or tissues affixed to a sl ide.
  • Nucleic acid ampl ification increases the copy number of sequences of interest such as the target nucleic acid.
  • amplification methods include the polymerase chain reaction method (PCR), the ligase chain reaction (LCR), self sustained sequence replication (3SR), nucleic acid sequence-based amplification (NASBA), the use of Q Beta replicase, reverse transcription, nick translation, and the like.
  • PCR polymerase chain reaction
  • LCR ligase chain reaction
  • NASBA nucleic acid sequence-based amplification
  • Q Beta replicase reverse transcription, nick translation, and the like.
  • the first cycle of ampl ification forms a primer extension product complementary to the target nucleic acid.
  • the target nucleic acid is single stranded RNA
  • a polymerase with reverse transcriptase activity is used in the first amplification to reverse transcribe the RNA to DNA, and additional amplification cycles can be performed to copy the primer extension products.
  • each primer must hybridize so that its 3 ' nucleotide is paired to a nucleotide in its complementary template strand that is located 3 ' from the 3' nucleotide of the primer used to replicate that complementary template strand in the PCR.
  • the target nucleic acid can be amplified by contacting one or more strands of the target nucleic acid with a primer and a polymerase having suitable activity to extend the primer and copy the target nucleic acid to produce a full length complementary nucleic acid or a smaller portion thereof.
  • Any enzyme having a polymerase activity that can copy the target nucleic acid can be used, including DNA polymerases, RNA polymerases, reverse transcriptases, and enzymes having more than one type of polymerase activity, and the enzyme can be thermolabile or thermostable. Mixtures of enzymes can also be used.
  • Exemplary enzymes include: DNA polymerases such as DNA Polymerase I ("Pol 1"), the
  • RNA polymerases such as E. coli, SP6, T3 and T7 RNA polymerases
  • reverse transcriptases such as AMV, M MuLV, MMLV, RNAse H' MMLV (Superscript®), Superscript® I I, ThermoScript®, HI V 1 , and RAV2 reverse transcriptases. All of these enzymes are commercially available.
  • Exemplary polymerases with multiple specificities include RAV2 and Tli (exo) polymerases.
  • Exemplary thermostable polymerases include Tub, Taq, Tth, Pfx, Pfu, Tsp, Tfl, Tli mdPyrococcus sp. GB D DNA polymerases.
  • Suitable reaction conditions are chosen to permit amplification of the target nucleic acid, including pH, buffer, ionic strength, presence and concentration of one or more salts, presence and concentration of reactants and cofactors such as nucleotides and magnesium and/or other metal ions (e.g., manganese), optional cosolvents, temperature, and thermal cycling profi le for amplification schemes comprising a polymerase chain reaction, and may depend in part on the polymerase being used as well as the nature of the sample.
  • Cosolvents include formamide (typically at from about 2 to about 10%).
  • glycerol typically at from about 5 to about 10%
  • DMSO typically at from about 0.9 to about 1 0%.
  • Techniques may be used in the amplification scheme in order to minimize the production of false positives or artifacts produced during amplification. These include ""touchdown" PCR, hot start techniques, use of nested primers, or designing PCR primers so that they form stem-loop structures in the event of primer-dimer formation and thus are not amplified.
  • Techniques to accelerate PCR can be used, for example, centrifugal PCR, which al lows for greater convection within the sample, and comprising infrared heating steps for rapid heating and cooling of the sample. One or more cycles of amplification can be performed.
  • An excess of one primer can be used to produce an excess of one primer extension product during PCR; preferably, the primer extension product produced in excess is the ampl ification product to be detected.
  • a plurality of different primers may be used to amplify different target nucleic acids or different regions of a particular target nucleic acid within the sample.
  • Amplified target nucleic acids may be subjected to post amplification treatments. For example, in some cases, it may be desirable to fragment the target nucleic acid prior to hybridization in order to provide segments which are more readily accessible. Fragmentation of the nucleic acids can be carried out by any method producing fragments of a size useful in the assay being performed; suitable physical, chemical and enzymatic methods are known in the art.
  • An amplification reaction can be performed under conditions which allow a nucleic acid associated with the optical sensing site to hybridize to the ampl ification product during at least part of an amplification cycle.
  • the assay is performed in this manner, real time detection of this hybridization event can take place by monitoring for light emission during amplification.
  • Real time PCR product analysis provides a well-known technique for real time PCR monitoring that has been used in a variety of contexts, which can be adapted for use with the methods described herein (see, Laurendeau et al. ( 1999) "TaqMan PCR-based gene dosage assay for predictive testing in individuals from a cancer family with INK4 locus haploinsufficiency" Clin Chem 45(7):982-6; Bieche et al . ( 1999) "Quantitation of MYC gene expression in sporadic breast tumors with a real-time reverse transcription-PCR assay" Cancer
  • Immunoassays can be conducted on the detection system of the invention, for example, at one or more optical sensing site of the system.
  • Suitable immunoassay systems include but are not limited to competitive and noncompetitive assay systems.
  • Such assay systems are typically used with techniques such as western blots, radioimmunoassays, EIA (enzyme immunoassay), ELISA (enzyme-linked immunosorbent assay), "sandwich” immunoassays, immunoprecipitation assays, precipitin reactions, gel diffusion precipitin reactions, immunodiffusion assays, agglutination assays, complement-fixation assays, immunoradiometric assays, fluorescent immunoassays, protein A immunoassays, and cellular immunostaining (fixed or native) assays to name but a few.
  • Immunoassay techniques particularly useful with the detection systems described herein include but are not limited to ELISA, "sandwich” immunoassays, and fluorescent immunoassays. Exemplary immunoassays are described briefly below (but are not intended by way of l imitation).
  • ELISAs generally involve preparing antigen, coating a wel l (e.g., an optical sensing site of the detection system) with the antigen, adding the antibody of interest conjugated to a detectable compound such as an enzymatic substrate (e.g., horseradish peroxidase or alkaline phosphatase) to the well and incubating for a period of time, and detecting the presence of the antigen.
  • a detectable compound such as an enzymatic substrate (e.g., horseradish peroxidase or alkaline phosphatase)
  • a detectable compound such as an enzymatic substrate (e.g., horseradish peroxidase or alkaline phosphatase)
  • a second antibody conjugated to a detectable compound may be added following the addition of the antigen of interest to the coated well.
  • a detectable compound may be added following the addition of the antigen of interest to the coated well.
  • a sample contains an unknown amount of biologically active analyte to be measured, which may be, for example, a protein.
  • the analyte may also be termed an antigen.
  • the sample may be spiked with a known or fixed amount of labeled analyte.
  • the spiked sample is then incubated with an antibody that binds to the analyte, so that the analyte in the sample and the labeled analyte added to the sample compete for binding to the available antibody binding sites. More or less of the labeled analyte will be able to bind to the antibody binding sites, depending on the relative concentration of the unlabeled analyte present in the sample.
  • the amount of labeled analyte bound to the antibody is measured, it is inversely proportional to the amount of unlabeled analyte in the sample.
  • the amount of analyte in the original sample may then be calculated based on the amount of labeled analyte measured, using standard techniques in the art.
  • an antibody that binds to a biologically active analyte may be coupled with or conjugated with a ligand, wherein the ligand binds to an additional antibody added to the sample being tested.
  • a ligand includes fluorescein.
  • the additional antibody may be bound to a solid support (e.g., an optical sensing site of the detection system).
  • the additional antibody binds to the ligand coupled with the antibody that binds in turn to the analyte or alternatively to the labeled analyte, forming a mass complex which allows isolation and measurement of the signal generated by the label coupled with the labeled analyte.
  • the biologically active analyte to be measured may be bound to a solid support (e.g., an optical sensing site of the detection system), and incubated with both an antibody that binds to the analyte and a sample containing the analyte to be measured.
  • the antibody binds to either the analyte bound to the solid support or to the analyte in the sample, in relative proportions depending on the concentration of the analyte in the sample.
  • the antibody that binds to the analyte bound to the solid support is then bound to another antibody, such as anti-mouse IgG, that is coupled to a label .
  • labels are detectable by spectroscopic, photochemical, biochemical, immunochemical, or chemical means.
  • useful nucleic acid labels include fluorescent dyes, enzymes, biotin, dioxigenin, or haptens and proteins for which antisera or monoclonal antibodies are avai lable.
  • Labeling agents optionally include, for example, monoclonal antibodies, polyclonal antibodies, proteins, or other polymers such as affinity matrices, carbohydrates or lipids. Detection proceeds by any of the methods described herein, for example, by detecting an optical signal in an optical waveguide.
  • a detectable moiety can be of any material having a detectable physical or chemical property.
  • Such detectable labels have been well-developed in the field of gel electrophoresis, column chromatography, solid substrates, spectroscopic techniques, and the like, and in general, labels useful in such methods can be applied to the present invention.
  • Preferred labels include labels that produce an optical signal.
  • a label includes without limitation any composition detectable by spectroscopic, photochemical, biochemical, immunochemical, electrical, optical, thermal, or chemical means.
  • the label is coupled directly or indirectly to a molecule to be detected such as a product, substrate, or enzyme, according to methods well known in the art.
  • a molecule to be detected such as a product, substrate, or enzyme
  • Non radioactive labels are often attached by indirect means.
  • a ligand molecule is covalently bound to a polymer. The ligand then binds to an anti ligand molecule which is either inherently detectable or covalently bound to a signal system, such as a detectable enzyme, a fluorescent compound, or a chemiluminescent compound.
  • ligands and anti-ligands can be used.
  • a ligand has a natural anti-ligand, for example, biotin, thyroxine, and Cortisol, it can be used in conjunction with labeled anti-ligands.
  • any haptenic or antigenic compound can be used in combination with an antibody.
  • the label can also be conjugated directly to signal generating compounds, for example, by conjugation with an enzyme or fluorophore.
  • Enzymes of interest as labels will primarily be hydrolases, particularly phosphatases, esterases and glycosidases, or oxidoreductases, particularly peroxidases.
  • Fluorescent compounds include fluorescein and its derivatives, rhodamine and its derivatives, dansyl, and umbell iferone.
  • Chemi luminescent compounds include luciferin, and 2,3- dihydrophthalazinediones, such as luminol .
  • Methods of detecting labels are wel l known to those of ski l l in the art.
  • the label may be detected by exciting the tluoi chrome with the appropriate wavelength of light and detecting the resulting fluorescence by. for example, a detection system as described herein.
  • enzymatic labels are detected by providing appropriate substrates for the enzyme and detecting the resulting reaction product (e.g., a reaction product capable of producing a detectable optical signal).
  • the detectable signal may be provided by luminescent sources.
  • Luminescence is the term commonly used to refer to the emission of light from a substance for any reason other than a rise in its temperature.
  • atoms or molecules emit photons of electromagnetic energy (e.g., light) when they move from an "excited state” to a lower energy state (usually the ground state); this process is often referred to as “radioactive decay”.
  • electromagnetic energy e.g., light
  • the luminescence process is referred to as "photoluminescence”.
  • the exciting cause is an electron
  • the luminescence process is referred to as "electroluminescence”.
  • electroluminescence results from the direct injection and removal of electrons to form an electron-hole pair, and subsequent recombination of the electron-hole pair to emit a photon.
  • Luminescence which results from a chemical reaction is usually referred to as “chemiluminescence”.
  • Luminescence produced by a living organism is usually referred to as “bioluminescence”.
  • photoluminescence is the result of a spin allowed transition (e.g., a single-singlet transition, triplet-triplet transition)
  • the photoluminescence process is usually referred to as "fluorescence”.
  • fluorescence emissions do not persist after the exciting cause is removed as a result of short-lived excited states which may rapidly relax through such spin allowed transitions.
  • photoluminescence is the result of a spin forbidden transition (e.g., a triplet- singlet transition)
  • the photoluminescence process is usually referred to as "phosphorescence”.
  • phosphorescence emissions persist long after the exciting cause is removed as a result of long-lived excited states which may relax only through such spin-forbidden transitions.
  • a “luminescent label” may have any one of the above-described properties.
  • Suitable chemiluminescent sources include a compound which becomes electronically excited by a chemical reaction and may then emit light which serves as the detectible signal or donates energy to a fluorescent acceptor.
  • a diverse number of families of compounds have been found to provide chemiluminescence under a variety of conditions.
  • One family of compounds is 2,3-dihydro- l ,4- phthalazinedione.
  • a frequently used compound is luminol, which is a 5-amino compound.
  • Other members of the family include the 5-amino-6,7,8-trimethoxy- and the dimethylamino[ca]benz analog. These compounds can be made to luminesce with alkaline hydrogen peroxide or calcium hypochlorite and base.
  • Chemi luminescent analogs include para-dimethyl ami no and -methoxy substituents. Chemiluminescence may also be obtained with oxalates, usually oxalyl active esters, for example, p-nitrophenyl and a peroxide such as hydrogen peroxide, under basic conditions. Other useful chemiluminescent compounds that are also known include— -alkyl acridinum esters and dioxetanes. Alternatively, luciferins may be used in conj unction with luciferase or lucigenins to provide
  • the present invention provides a method of monitoring one or more pharmacological parameter, for example, pharmacodynamic (PD) and/or pharmacokinetic (PK) parameters, useful for assessing efficacy and/or toxicity of a therapeutic agent.
  • the method comprises subjecting a sample of bodily fluid from a subject administered with the therapeutic agent to a detection device for monitoring the one or more pharmacological parameter; using the detection device as described herein to yield detectable signals indicative of the values of the more than one pharmacological parameter from the sample; and detecting the detectable signal generated from said sample of bodily fluid.
  • the samples tested can include a large number of a variety of small molecules (e.g., screening libraries) which are of interest when investigating new drugs. Accordingly, the detection system described herein is useful for screening libraries of small molecules to investigate their ability to interact with certain biologically active analytes to reveal potential new drugs. Further screening of some or all small molecule candidates may reveal adverse drug effects and toxicity.
  • small molecules e.g., screening libraries
  • the samples can include molecules which are tested for toxicity.
  • the scanning light source moves through its scanning path to a point at which it is coupled to and in optical communication with one or more in-coupling or excitation waveguides, thus generating a pulse of light within the waveguides.
  • the light travels along the waveguides, reaches the optical sensing sites and interacts through the sensor, for example, an optical transducer.
  • the samples are positioned at or near the waveguides.
  • the secondary light leaving the sensor couples into the out-coupling or collection waveguides and travels down the waveguide to its end at an edge of the substrate, for example, a chip facet. Light exiting the out-coupling or collection waveguides is then detected by the different elements of the detector, which can be a detector array.
  • the substrate comprises a plurality of waveguides that serve as both in/out-coupling waveguides, with the light source and the detector being coupled to and in optical communication with opposite ends of the waveguides.
  • the in/out-coupl ing waveguides collect light from the light source and guide the secondary light to the detector through one or more adapter.
  • the scanning light source/detector moves through its scanning path to a point at which the light source is coupled to and in optical communication with one or more in- coupling waveguides, thus generating a pulse of light within the waveguides.
  • the detector is coupled to and in optical communication with one or more out-coupling waveguides.
  • the l ight travels along the waveguides, reaches the optical sensing sites and interacts through the sensor, for example, an optical transducer.
  • the samples are positioned at or near the waveguides.
  • the secondary light leaving the sensor couples into the out-coupling waveguides and travels down the waveguide to its end at an edge of the substrate, for example, a chip facet.
  • the substrate comprises a plural ity of waveguides that serve as both in/out-coupling waveguides
  • the light source/detector chip comprises in-coupling and out-coupling waveguides that are coupled through at least one combiner.
  • the light waves generated by the light source elements are coupled into the in-coupl ing waveguides of the l ight source/detector chip, then combined by combiners into the out-coupling waveguides, which couple this primary light to the waveguides of the substrate.
  • the secondary l ight wave leaving the sensor travels through the same waveguides of the substrate to out- coupling waveguides of the light source/detector chip which guide the light wave to the detector elements.
  • detection of a sample includes delivering a sample suspected of containing a target to be detected to an optical sensing site of the detection system.
  • Delivering a sample to the system can include pipetting of a fluid to the optical sensing site.
  • Other delivery means can include but are not limited to a robotic fluid delivery system or physically depositing a non-fluid or semi-fluid sample at the optical sensing site, either by hand or with the aid of a tool or robot manipulation system.
  • a first light wave produced by the scanning light source is provided to one or more of the plurality of waveguides in optical communication with the optical sensing site.
  • the first light wave is transduced (e.g., measurably changed) by the sensor associated with the optical sensing site to form a second light wave carried back in one or more of the plurality of out-coupling or collection waveguides which are in optical communication with the optical sensing site.
  • a measurable change in the second light wave is detected using the detector which is in optical communication with the out-coupling or col lection waveguides. Detection of a measurable change in the second light wave indicates that the sensor has interacted with the target.
  • the waveguides described herein can be arranged substantially parallel as illustrated general ly in the accompanying figures.
  • the detection method includes generating one or more light wave by the scanning light source which couples into the substrate at some point along its scanning path to produce the first light wave in one or more of the waveguides in a controlled manner.
  • the different light source elements of the scanning light source can be switched on simultaneously to generate one or more input light waves.
  • the plural ity of light waves can be coupled into the substrate to controllably produce the first light wave in one or more of the waveguides.
  • all in-coupling waveguides are provided with a first light wave and simultaneous detection of second light waves at each out-coupling waveguide is achieved using a detector that is a photodetector array.
  • each waveguide can be individually addressed with a first light wave.
  • the order of addressing the waveguides can be sequential, staggered, random or in any order desired. Rapid scanning of the entire array of optical sensing sites can be achieved with the aid of the photodetector array since any second light wave associated with each out- coupling waveguide can be simultaneously detected.
  • a single excitation waveguide is provided with a first l ight wave and simultaneous detection of second l ight waves at each col lection waveguide is achieved using a detector that is a photodetector array.
  • the two-dimensional waveguide array is configured as an array of 128 excitation waveguides and 128 collection waveguides, then it would be possible to simultaneously detect second light waves (if any) generated from 1 28 optical sensor sites after providing a single first lightwave in a first excitation waveguide.
  • 128 optical sensing sites can be interrogated for presence or absence of target simultaneously.
  • a second excitation waveguide can be provided thereby triggering the interrogation of a second set of 128 optical sensing sites. The process can rapidly be repeated until every excitation waveguide has been excited and the entire array of optical sensing sites have been interrogated.
  • the method of using the detection system involves the detection of a substance, including but not limited to a biologically active analyte including a nucleic acid, a protein, an antigen, an antibody, a panel of proteins, a microorganism, a gas, a chemical agent and a pollutant.
  • a single nucleotide polymorphism is detected in the target.
  • expression of a gene is detected upon detection of the target.
  • ddNTPs dideoxynucleotriphosphates
  • Identification of the single base added to the 3' end of the probe molecule can be done in one of three ways: parallel channels for each of the four bases using a different labeled ddNTP in each channel ; sequential SBEX reactions using a different labeled ddNTP in each reaction; or wavelength discrimination of the four possibil ities using a different fluorescent label for each ddNTP.
  • the first of these methods may be preferred.
  • SBEX may be used in oligonucleotide genotyping and SNP detection systems, and is advantageous over traditional hybridization assays, for example, due to greater base specificity, production of a covalent bond between the labeled dNTP and the probe, and simultaneous detection of multiple bases.
  • polymorphisms can be done with ease. By patterning the waveguide with different capture sequences, different points in a sequence, for example, a genome, a chromosome and/or a gene, may be assayed. As SBEX only requires a fluorescent label on the ddNTP monomers used, all instances of a particular base wi ll be detected. In order to do the same thing with a traditional DNA hybridization assay, each probe DNA for each capture sequence would have to be fluorescently labeled.
  • the enzyme-catalyzed reaction has two distinct advantages. First, a stable covalent bond forms between the stationary phase and a labeled monomer, e.g., a Cy5-labeled monomer. This increases the assay sensitivity versus traditional hybridization assays where the fluorescent label is captured by the stationary phase via non-covalent interactions (duplex formation). Optionally a stringent washing step can be employed. Second, the polymerase enzyme incorporates the dideoxynucleotide with high fidelit— due to the replication accuracy of a polymerase, in general only the base that is complementary to the target base will react. SBEX is particularly well suited for planar waveguide technology, benefiting from the increased speed of a washless assay and increased sensitivity provided by kinetic data.
  • a labeled monomer e.g., a Cy5-labeled monomer.
  • a stringent washing step can be employed.
  • the polymerase enzyme incorporates the dideoxynucleotide with high fidelit—
  • Fluorescence imaging is sensitive to speed, sensitivity, noise and resolution, and each may be optimized for use in the invention; for example, speed may be increased to decrease assay times.
  • Base extension may be detected using a CCD camera, a streak camera, spectrofluorometers, fluorescence scanners, or other known fluorescence detection devices, which generally comprise four elements: an excitation source; a fluorophore; a filter to separate emission and excitation photons; and a detector to register emission photons and produce a recordable output, typically an electrical or photographic output.
  • thermostable polymerases such as pfu, Taq, Bst, Tfl, Tgo and Tth polymerase, DNA
  • the polymerase may be a DNA-dependent DNA polymerase, a DNA-dependent RNA polymerase, a RNA-dependent RNA polymerase, a RNA- dependent DNA polymerase or a mixture thereof, depending on the template, primer and NTP used.
  • the polymerase may or may not have proofreading activity (3 ' exonuclease activity) and/or 5' exonuclease activity).
  • the capture molecule and/or the analyte molecule of the invention may be any nucleic acid, including, but not limited to, DNA and/or RNA and modifications thereto known in the art, and may incorporate 5'-0-( l -thio)nucleoside analog triphosphates, . alpha. -thiotriphosphate, 7-Deaza-.alpha.- thiotriphosphate, N6-Me-.alpha. -thiotriphosphate, 2'-0-Methyl-triphosphates, morpholino, PNA, aminoalkyl analogs, and/or phosphorothioate.
  • immunoassays can be used with the present method of using the detection system.
  • the optical sensing site of the detection system of the invention can be adapted to support an immunoassay, for example, by including one or more immunoassay reagent at or withi n the optical sensing site.
  • an interaction between the optical sensing site and a sample being tested for a biologically active analyte can include an immunoassay conducted at the optical sensing site.
  • the optical sensing site interacting with the biologically active analyte can include an outcome of an immunoassay. In this manner, presence or absence of the analyte can be determined. Additional ly the amount of analyte can be quantified.
  • the immunoassay supported is a fluorescent assay. It is envisioned that the immunoassay can be a competitive or non-competitive immunoassay.
  • the immunoassay supported is an EL1SA.
  • the optical detection system may be used in a range of applications including biomedical and genetic research as well as clinical diagnostics.
  • Arrays of polymers such as nucleic acids may be screened for specific binding to a target, such as a complementary nucleotide, for example, in screening studies for determination of binding affinity and in diagnostic assays.
  • sequencing of polynucleotides can be conducted, as disclosed in U.S. Pat. No. 5,547,839.
  • the nucleic acid arrays may be used in many other appl ications including detection of genetic diseases such as cystic fibrosis or diagnosis of diseases such as HIV, as disclosed in U.S. Pat. No. 6,027,880 and U.S. Pat. No. 5,861 ,242.
  • Genetic mutations may be detected by sequencing or by hydridization.
  • genetic markers may be sequenced and mapped using Type-IIs restriction endonucleases as disclosed in U.S. Pat. No. 5,710,000.
  • Gene expression may be monitored by hybridization of large numbers of mRNAs in parallel using high density arrays of nucleic acids in cells, such as in microorganisms such as yeast, as described in Lockhart et a! ., Nature Biotechnology, 14: 1675-1 680 ( 1996).
  • Bacterial transcript imaging by hybridization of total RNA to nucleic acid arrays may be conducted as described in Saizieu et al., Nature Biotechnology, 16:45-48 ( 1998). Accessing genetic information using high density DNA arrays is further described in Chee, Science 274:610-614 ( 1996).
  • optical detection systems of the invention may be used in combination with protein and chemical microarrays, including arrays of protei ns, antibodies, small molecule compounds, peptides, and carbohydrates, or cell or tissue arrays, as described for example in Xu, Q. and Lam, K. S., J. Biomed. Biotechnol. 5:257-266 (2003).
  • Protein and chemical arrays may be used in combination with the methods and devices of the invention in a number of potential applications, including but not l imited to proteomics (including assays of both protein-protein interactions and protein-l igand interactions), screening assays for drug discovery, and toxicology testing. In some appl ications these assays may uti l ize label-free optical sensing methods as described, for example, in U.S. Pat. No. 7,349,080 and U.S. Pat. No. 7,292,336.
  • a further potential application is detection of chemical and/or biological warfare agents, including but not limited to bacterial spores (for example, as descri bed in U .S. Pat. No. 6,498.04 1 ): bacterial agents (e.g., Bacillus anthracis, Yersinia pest is, F.
  • bacterial agents e.g., Bacillus anthracis, Yersinia pest is, F.
  • titlararensis Bruce/la, Clostridium Botulinum, Clostridium tetani, Coxiella burnetii, and Vibrio cholerae); viral agents (e.g., variola virus, viral encephalitis agents such as Venezuelan equine encephalitis, western equine encephal itis and eastern equine encephalitis; and viral hemorraghic fever agents such as arenaviridae, bunyaviridae, filoviridae, and flaviviridae) and toxins (e.g., Staphylococcus enterotoxin B, botuhnum toxin, ricin, and mycotoxins, as well as anticrop agents (e.g., Pitccinia graministrititi, Piricularia oryzae, Tileltia caries, Tilettia foetida, Fusarium fungus, and herbicides).
  • viral agents e.g.
  • Bacterial and viral agents may be detected using, for example, nucleic acid based methods such as real-time PCR, antibody-based detection methods such as ELISA, or using antimicrobial peptides as described by Kulagina et al., Sens. Actuators B. Chem. 121 : 1 50- ] 57 (2007).
  • Non-limiting potential applications include detection and diagnosis of viral and bacterial infectious diseases (e.g. AIDS, Bird Flu, SARS, West Nile virus); point-of-care monitoring of patients (e.g, detection of sugar and insul in levels in diabetic patients, detection of blood gas levels or lactic acid levels); pregnancy testing; detection of drugs or narcotics (e.g., cocaine, ecstasy,
  • viral and bacterial infectious diseases e.g. AIDS, Bird Flu, SARS, West Nile virus
  • point-of-care monitoring of patients e.g, detection of sugar and insul in levels in diabetic patients, detection of blood gas levels or lactic acid levels
  • pregnancy testing detection of drugs or narcotics (e.g., cocaine, ecstasy,
  • methamphetamines opiates
  • detection of chemical and explosive substances e.g, RDX, TNT, nitroglycerin
  • environmental monitoring of air, water or soil samples including the detection of pesticides, heavy metals, nitrates or phosphates.
  • the working system described here can also be a sub-system within a much larger bio- analysis system.
  • the bio-analysis system could include all the aspects of sample preparation prior to optical detection, the post processing of data collected in the optical detection phase and finally decision making based on these results.
  • Sample preparation may include steps such as: extraction of the sample from the tested subject (human, animal, plant environment etc.); separation of different parts of the sample to achieve higher concentration and purity of the molecules under investigation; sample amplification (e.g. through PCR); attachment of fluorescence tags or markers to different parts of the sample; and spotting of the sample onto the substrate.
  • the post processing of the collected data may include: normalization; background and noise reduction; and statistical analysis such as averaging over repeated tests or correlation between different tests.
  • the decision making may include: testing against a predefined set of rules and comparison to information stored in external databases.
  • a method of diagnosing a disease comprises reviewing or analyzing data relating to the presence and/or the concentration level of a target i n a sample.
  • a concl usion based upon review or analysis of the data can be provided to a patient, a health care provider or a health care manager.
  • the conclusion is based upon the review or analysis of data regarding a disease diagnosis. It is envisioned in another embodiment that providing a conclusion to a patient, a health care provider or a health care manager includes transmission of the data over a network.
  • One aspect of the invention is a business method comprising screening patient test samples for the presence or absence of a biological ly active analyte to produce data regarding the analyte, collecting the analyte data, and providing the analyte data to a patient, a health care provider or a health care manager for making a conclusion based upon review or analysis of the data regarding a disease diagnosis.
  • providing a conclusion to a patient, a health care provider or a health care manager includes transmission of the data over a network.
  • FIG. 15 is a block diagram showing a representative example of a logic device through which reviewing or analyzing data relating to the present invention can be achieved. Such data can be in relation to a disease, disorder or condition in an individual.
  • FIG. 15 shows a computer system (or digital device) 1500 connected to an apparatus 1 520 for use with the detection system 1524 to, for example, produce a result.
  • the computer system 1 500 may be understood as a logical apparatus that can read instructions from media 151 1 and/or network port 1505, which can optional ly be connected to server 1 509 having fixed media 1 512.
  • the system shown in FIG. 1 5 includes CPU 1 501 , disk drives 1503, optional input devices such as keyboard 1515 and/or mouse 1 516 and optional monitor 1507.
  • Data communication can be achieved through the indicated communication medium to a server 1 509 at a local or a remote location.
  • the communication medium can incl ude any means of transmitting and/or receiving data.
  • the communication medium can be a network connection, a wireless connection or an internet connection. Such a connection can provide for communication over the World Wide Web. It is envisioned that data relating to the present invention can be transmitted over such networks or connections for reception and/or review by a party 1 522.
  • the receiving party 1 522 can be but is not limited to a patient, a health care provider or a health care manager.
  • a computer-readable medium includes a medium suitable for transmission of a result of an analysis of an environmental or biological sample.
  • the medium can include a result regarding a disease condition or state of a subject, wherein such a result is derived using the methods described herein.
  • a kit comprises detection system as described herein and reagents for detecti ng a target in the sample.
  • the kit may optionally contain one or more of the fol lowing: one or more fluorescent or l uminescent molecular tag, and one or more biologically active analyte incl uding a nucleic acid, protein, microorganism or chemical agent.
  • kits can be retained by a housing. Instructions for using the kit to perform a described method can be provided with the housing, and can be provided in any fixed medium.
  • the instructions may be located inside the housing or outside the housing, and may be printed on the interior or exterior of any surface forming the housing that renders the instructions legible.
  • a kit may be in multiplex form for detection of one or more different target biologically active analytc including nucleic acid, protein, microorganism, gas, chemical agent or pollutant.
  • a kit 1603 can include a detection system 1600, a housing or container 1 602 for housing various components. As shown in FIG. 16, and described herein, the kit 1603 can optionally include instructions 1601 and reagents 1 605, for example, DNA hybridization or immunoassay reagents. Other embodiments of the kit 1603 are envisioned wherein the components include various additional features described herein.
  • a kit for assaying a sample for a target includes a detection system including a scanning light source, a detector, and a substrate.
  • the substrate can include a plural ity of excitation waveguides and a plurality of collection waveguides as described herein.
  • the excitation waveguides and collection waveguides of the substrate cross or intersect to form intersection regions and a two-dimensional array.
  • the system further includes a plural ity of optical sensing sites.
  • the optical sensing sites are in optical communication with one or more excitation waveguides and one or more collection waveguides.
  • the kit further includes packaging and instructions for use of the system.
  • the crossing of the excitation waveguides and collection waveguides is substantially perpendicular.
  • a kit for assaying a sample for a target includes a detection system including a scanning light source, a detector, and a substrate.
  • the substrate can include a plurality of substantially parallel in-coupling waveguides and a plurality of substantially parallel out-coupling waveguides as described herein.
  • the system can further include a plurality of optical sensing sites. The optical sensing sites are in optical communication with one or more waveguides.
  • the kit further includes packaging and instructions for use of the system.
  • the kit includes a detection system that is a planar l ightwave circuit (PLC).
  • PLC planar l ightwave circuit
  • a substrate for assaying a sample for a target is provided.
  • the substrate is a PLC.
  • the starting material for manufacturing PLC devices is a wafer usual ly made of silicon (Si) or sil ica (Si02). The most common wafer diameters in use are 4". 6" and 8".
  • the manufacturing process for PLC devices involves two basic processes, namely, deposition and etching. A short description of each of them is given below.
  • the methods of manufacturing the systems described herein can include, but are not limited to laser writing, UV writing and photonic band-gap waveguide methods.
  • the manufacturing process in some embodiments includes one or more steps of deposition, masking and etching.
  • Deposition [000389] In the deposition step a layer of well defined material having well controlled thickness is deposited across the entire wafer.
  • the most common materials used for waveguide layer deposition are silica (Si02), also known as glass, and silicon nitride (Si3N4).
  • the optical properties of the silica (mainly its refractive index) is control led by the amount of doping (Ge, P, and B etc.) introduced during the deposition.
  • Other materials such as silicon, glass, epoxy, lithium niobate, indium phosphide and SiON (silicon oxynitride) and its derivatives are also used.
  • materials can include but are not limited to si licon, si lica (Si02), glass, epoxy, lithi um niobate and indium phosphide.
  • the deposition step is done using several technologies such as PECVD (Plasma- Enhanced Chemical Vapor Deposition), LPCVD (Low Pressure CVD), APCVD (Atmospheric pressure CVD), FHD (Flame Hydrolysis Deposition) and others well known in the art.
  • PECVD Pulsma- Enhanced Chemical Vapor Deposition
  • LPCVD Low Pressure CVD
  • APCVD Admospheric pressure CVD
  • FHD Flume Hydrolysis Deposition
  • FIG. 17A illustrates an exemplary substrate 1 704 as a schematic structure created after two consecutive deposition steps of a cladding 1721 layer and a core 1723 layer over a silicon 1726 layer, which can be a wafer.
  • these two layers differ in the refraction index which is achieved by using different levels of doping.
  • Typical thicknesses for the different layers are: Cladding up to about 20 ⁇ and core up to 6 ⁇ .
  • the thickness of the silicon 1 726 wafer can range from about 0.5 mm to 1 mm.
  • the desired two-dimensional structure of the PLC device is transferred to the deposited wafer by masking the areas not to be etched away.
  • the masking is done in several steps involving covering the wafer with light sensitive material, exposing it to light through l ithographic masks and removing the exposed material leaving in place the mask. The result of such steps is shown in FIG. 17B where a mask 1 725 is shown on top of the core 1723 layer of the substrate 1704.
  • FIG. 17C shows the results of the etching step which results in a waveguide 1 727.
  • an over-cladding or top cladding 1 729 layer is created using a deposition step similar to the one described above.
  • the results are shown in FIG. 1 7D.
  • the resulting waveguide 1727 can be surrounded by a top cladding 1 729 and a cladding 1721 over a si licon 1726 layer.
  • CM P Chemical Mechanical Planarization
  • FIG. I 8 An exemplary simpl ified flow-chart of the manufacturing process is shown in FIG. I 8.
  • the present invention features methods and optical arrangements enabling the performance of two or more assays separated by time and / or separated by the color / wavelength of their emitted fluorescence on each and every sensing-well of a waveguide based bio-sensing chip.
  • at least one of the assays performed in each and every well is attempting to measure the signal generated by the presence of a target moiety.
  • the other assays measure the signal generated by one or more secondary moieties in the sample, either naturally occurring or intentionally added, or by moieties immobil ized to the sensing wel l, which signal indicates the level of a critical parameter such as temperature, excitation-light intensity, reagents aging, blood hematocrit and non-specific binding.
  • These signals generated by the secondary moieties can serve to calibrate and / or normalize the signal measured from the target moiety in order to achieve a more consistent and reliable assay result which takes into account the variation of these signal affecting parameters.
  • optical arrangement that can be used for the current invention may be based on the optical system as described above and further described in US 7,951 ,583 and US 8, 1 87,866 and can include in addition:
  • Interference filters allowing one or more of the fluorescent signals excited by one or more of the lasers to reach the optical detector at the output of the sensing chip while blocking all excitation light from the lasers.
  • Moieties labelled with different fluorescent tags which fluorescence can be excited using one or more of the excitation lasers and can be measured through one or more of the interference filters.
  • one or more specific capture moiet ies are immobilized in each of the sensing wel ls of the waveguide-based sensing chip.
  • the specific target moieties i n the sample start binding to the corresponding specific capture moieties in the sensing well.
  • Shinning laser light having a wavelength within the excitation spectrum of one or more of the fluorescent tags, generates fluorescence with wavelengths within the emission spectrum of these fluorescent tags.
  • This fluorescent l ight signal is captured in the sensing-wel l and directed to an optical detector through interference fi lters that ' fi lters out' most of the remaining laser l ight, thus allowing the measurement of only the desired fluorescent light.
  • a mix of two different capture molecules are immobil ized in each of the sensing wel ls.
  • the mix contains, at a given ratio (e.g. 90: 10), a capture antibody specific to the target being detected and a second capture antibody which is specific to a control / normal izing moiety.
  • a known concentration of the control / normalizing moiety is added to the sample, together with two detection antibodies, one specific to the target being detected and the other specific to the control / normalizing moiety.
  • the two detection antibodies are each fluorescently labelled with a different fluorescent tag, each tag emitting at a different fluorescence wavelength.
  • the binding to the surface of the sensing wells is monitored in given time-intervals (e.g. every 5 seconds) by shining laser-light from two different lasers, one after the other, into the excitation waveguides of the sensing chip.
  • time-intervals e.g. every 5 seconds
  • the second reading appears upon coupling laser #2 into the excitation waveguides and it is a measure of the fluorescence emitted by the labelled detection antibodies specific to the control / normalizing moiety.
  • the first reading is normal ized to the second reading by dividing one by the other or by implementing any other suitable mathematical algorithm, leading to the elimination or reduction of aberrations caused by factors (such as temperature changes and reagents aging) which simi larly affect the target and the control moiety.
  • the time interval using for the first laser-light may be different than the interval used for the second laser-light.
  • one of the fluorophores may be more susceptible to photo-bleaching than the other fluorophore, so longer time intervals can be used with the photo-sensitive fluorophore in order to reduce photo- bleaching.
  • a mix of two different capture molecules are immobi lized in the each of the sensing wel ls.
  • the mix contains, at a given ratio (e.g. 90: 10), a capture antibody specific to the target being detected and a second capture antibody which is specific to a control / normal izing moiety.
  • the test is performed in two steps. In the first step, a fluorescently label led detector antibody specific to the target is added to the sample at a known concentration and the mix is presented to the sensing chip. The binding to the surface of the sensing wells is monitored in given time- intervals (e.g. every 5 seconds) by shining laser-light from a single laser, into the excitation waveguides of the sensing chip.
  • the control / normal izing moiety together with its specific fluorescently labelled detector antibody are added to buffer or other selected matrix (e.g. serum, plasma, oral fluid) at a known concentration and the mix is presented to the sensing chip.
  • the binding to the surface of the sensi ng wel ls is monitored in given time-intervals (e.g. every 5 seconds) by shining laser- l ight from the same single laser, into the excitation waveguides of the sensing chip.
  • time-intervals e.g. every 5 seconds
  • the results of the first step are normal ized at the end of the second step by the results of the second step.
  • the normalization can be done by dividing point-by-point or by dividing each point of the first step by the average or slope (over time) of the second step, or by dividing the signal slope (over lime) of the first step by the slope (over time) of the second step or any other mathematical algorithm that helps in the elimination of aberrations caused by factors (such as temperature changes and reagents aging) which similarly affect the target and the control moiety.
  • a single capture molecule is immobilized in the each of the sensing wells.
  • the capture molecule is specific to the target being detected.
  • the test is performed in two steps.
  • a fluorescently labelled detector antibody specific to the target is added to the sample at a known concentration and the mix is presented to the sensing chip.
  • the binding to the surface of the sensing wells is monitored in given time-intervals (e.g. every 5 seconds) by shining laser-light from a single laser, into the excitation waveguides of the sensing chip.
  • time-intervals e.g. every 5 seconds
  • the target moiety together with its specific fluorescently labelled detector antibody are added to buffer or other selected matrix (e.g. serum, plasma, oral fluid) at a known concentration and the mix is presented to the sensing chip.
  • buffer or other selected matrix e.g. serum, plasma, oral fluid
  • the binding to the surface of the sensing wells is monitored in given time-intervals (e.g. every 5 seconds) by shining laser-light from the same single laser, into the excitation waveguides of the sensing chip.
  • time-intervals e.g. every 5 seconds
  • the normalization can be done by dividing point-by-point or by dividing each point of the first step by the slope (over time) of the second step, or any other mathematical algorithm that helps in the elimination of aberrations caused by factors (such as temperature changes and reagents aging).
  • a mix of three different capture molecules are immobil ized in the each of the sensing wells.
  • the mix contains, at a given ratio (e.g. 80: 10: 10), the capture antibody specific to the target being detected and a two other capture antibodies which are each specific to one control / normalizing moiety.
  • a known concentrations of the two control / normal izing moieties preferably one at low concentration and the other at high concentration, are added to the sample, together with three detection antibodies, one specific to the target being detected and the two other specific to the two control / normalizing moieties.
  • the low concentration and the high concentration can be chosen to span the entire range to be measured.
  • the low concentration can be within 10, 20, 30, 40, or 50% of the low end of the range to be measured
  • the high concentration can be within 10, 20, 30, 40, or 50% of the high end of the range to be measured.
  • the three detection ant ibodies are each fl uorescently labelled with a different fl uorescent tag.
  • each tag emitting at a di fferent fluorescence wavelength.
  • the binding to the surface of the sensing wel ls is monitored in given time-intervals (e.g. every 5 seconds) by shining laser-light from three different lasers, one after the other, into the excitation waveguides of the sensing chip.
  • time-intervals e.g. every 5 seconds
  • the first reading appears upon coupling laser # 1 into the excitation waveguides and it is a measure of the fluorescence emitted by the labelled detection antibodies specific to the target being detected.
  • the second reading appears upon coupling laser #2 into the excitation waveguides and it is a measure of the fluorescence emitted by the first labelled detection antibody specific to the first control / normalizing moiety.
  • the third reading appears upon coupl ing laser #3 into the excitation waveguides and it is a measure of the fluorescence emitted by the second label led detection antibody specific to the second control / normalizing moiety.
  • the signal from the two control / normal izing moieties are used for normalizing the first reading using a suitable mathematical algorithm.
  • the signal from the two control / normalizing moieties are used to generate a 'calibration curve / line' which is used to calculate the unknown concentration of the target.
  • the substrate 804 of the sensing chip as shown in FIG. 8, for example, can be incorporated into a cartridge with a fluid port for introducing a sample, a flow path for directing the sample to the substrate 804, and a waste reservoir for collecting the sample after is passes over the substrate, as further described in U. S. Patent Publication No. 20160033412, which is herein incorporated by reference in its entirety.
  • the detector 806 which can be an optical laser head.
  • the substrate can be modified to include a loop back waveguide 713.
  • a software and control system can be used to align the cartridge with the optical head.
  • a sensor detects the insertion of the cartridge and triggers the light source to send a signal to the substrate and begin the alignment process.
  • the sensor can be a mechanical, electrical, or optical switch that is triggered when the cartridge is inserted into the reader.
  • the light travels through the loop back waveguide and a detector, such as a CMOS detector, at the other end of the loop back waveguide 71 3 receives the light.
  • a detector such as a CMOS detector
  • the detector receives the light transmitted through the loopback waveguide. The amount of light received determines the status of the al ignment.
  • the system uses broad passes of the optical head to rapidly detect the location of the loopback channel .
  • the auto-al ignment software controls the X & Y axis motion of the optical head and repeats this process til l the al ignment is done.
  • a smart algorithm is developed to optimize the al ignment through X Y movement of the optical head ti l l complete al ignment happens.
  • Time outs can be put in place to detect error condit ions.
  • a home position can be provided to bring the head to a fixed location in case the process has to be restarted.
  • FIG. 23 i llustrates a flowchart of an embodiment of the automated alignment procedure that can be implemented through a control system and/or software in the reader.
  • a cartridge is inserted into a reader.
  • the reader system can ensure that the sample does not flow through the cartridge until after al ignment is successfully completed. This can be accomplished, for example, by not applying a vacuum or activating a pump until al ignment is successfully completed.
  • light is sent to one end of the loop back waveguide of the substrate.
  • a l ight detector at the other end of the loop back waveguide is used to measure the amount of light that is entering the loop back waveguide.
  • step 2306 if the light measured by the loop back waveguide detector is greater than a predetermined level or at a maximum, the alignment is complete 2308. If the light measured by the detector is less than the predetermined level or less than a maximum detected level, then the optica! head that is providing the light to the substrate is moved in the X and/or the Y direction by a predetermined amount, which may be on the order of microns, as shown in step 23 10. The procedure then returns to step 2304, where light is again sent to the loop back waveguide and measured with the detector.
  • the optical head can be moved along a single axis, such as the X direction, until alignment is achieved along the X axis, and then the optical head can be moved along the second axis, such as the Y axis, until alignment is achieved along the Y axis.
  • the increase or decrease in light measured by the detector as the optical head is moved in a particular direction can be used to determine whether the optical head is being moved in the correct direction for achieving alignment. An increase in measured light indicates that the movement of the optical head is proceeding in the correct direction, whi le a decrease in measured light indicates that the movement of the optical head is proceeding in the wrong direction.
  • the optical head can be alternately moved in the X and Y directions during alignment.
  • FIG. 24 i llustrates a flowchart of the antibody printing procedure.
  • step 2400 droplets of antibodies are disposed onto a substrate using a plurality of droppers arranged in array.
  • the array is l inear.
  • the array can be two dimensional.
  • the droppers can dispose the droplets in a non-contact manner, meaning that the droplets are disposed above the surface of the substrate and then fall onto the substrate. The droppers never contact the substrate.
  • a high resolution and high precision image of the droplets on the substrate can be automatical ly taken.
  • step 2404 image processing software can be used to analyze the image to determine the size, volume, shape, and location of the droplets which can be used in step 2406 to determine whether all the antibody droplets are placed correctly on the substrate. If the droplets are placed correctly, then the substrate can be kept for use and further processing in step 2408. If the droplets are not placed correctly, then the substrate can be thrown away in step 2410.
  • FIGS. 25A-25D illustrate a linear array of droppers 2500 that are dispensing droplets 2502 onto a substrate 2504.
  • the linear array of droppers are part of a specialized printer 2506 and imager.
  • One feature of the sensors of the current invention is the ability to detect low numbers of biologically active analytc molecules.
  • the following experiments demonstrate this capability.
  • the limit of detection (LOD) of the sensors was determined by measuring fluorescently labeled ovalbumin in solution. The dye used was Alexa Fluor 660; it was excited at 658 nm and emission was measured at 690 nm. All measurements were made at room temperature (25° C). LOD was assayed using two different methods: 1 ) determination of the lowest concentration of labeled oval bumin detectable above buffer background with better than 95% confidence (P ⁇ 0.05, Student's t-test); and 2) determination of the analytical sensitivity from a standard curve of sensor response vs.
  • the number of molecules detected per sample was calculated as follows.
  • the sample volume used in all experiments was 50 microliters. Thus, at a concentration of 0.1 pM for example, there were 3 x l 0 6 molecules per sample. Since all molecules were evenly distributed across the entire volume, the number of molecules available per sensor was ⁇ 1000. The thickness of the fluid containing this number of molecules was ⁇ 1 .7 mm. Using the diffusion parameters, it can be estimated that 0.1 % to 1 % of these molecules were able to bind to the surface of each sensor, or 1 to 10 molecules.
  • the present invention is useful for measuring protein-protein interactions.
  • the waveguide-based optical detection technology was used to measure the binding of ovalbumin to anti-ovalbumin antibodies.
  • chips with 52.5x4,500 pm sensing elements were dip-coated with anti-ovalbumin antibodies. This procedure involved several steps. First, the chips were cleaned. A chip fabricator coated the active surface of the chips with a thin polymer layer to protect it during wafer dicing. The polymer layer was removed by immersing chips in acetone for 5 minutes, followed by isopropanol for 5 minutes and a second isopropanol rinse for an additional minute. Chips were then washed in deionized water three times and dried in a vacuum desiccator.
  • Chips were immersed in a Piranha solution containing 9% (v/v) H202 and 66.5% (v/v) H2S04 for 45 minutes with agitation. This step removed organic contaminants and produced a thin ( 1 -2 nanometer) layer of reactive silanol groups that were used for coupling antibodies. Chips were then rinsed four times in deionized water, followed by a final rinse in doubled distil led water.
  • biotinylated anti- ovalbumin antibody e.g. US Biological, polyclonal
  • ovalbumin To assay the interaction of ovalbumin with the anti-ovalbumin antibodies, fluorescently labeled ovalbumin in a solution containing phosphate buffered saline, pH 7.4, and 0.1 mg/mL bovine serum albumin, was added to a sample well on the surface of a chip as depicted in FIG. 7D. An ovalbumin concentration range of four orders of magnitude (from 0.1 pM to 1 ,000 pM) was used. The kinetics of the binding reaction were then monitored over a period of 10 minutes at room temperature, and fluorescence intensity was measured at 690 nm.
  • FIG. 19 shows the quantitative sensor response for the binding reaction. Fluorescence intensity at each ovalbumin concentration used is indicated with plot symbols and error bars and is plotted on a graph with log based abscissa and ordinate axes. The solid line in FIG. 19 is a curve fit.
  • the waveguide-based optical detection system can be used to quantify the polymerization of DNA in a primer extension assay.
  • a primer extension reaction an mRNA target hybridizes to a DNA primer, and reverse transcriptase uses the mRNA target as a template to add deoxynucleotides (dATP, dTTP. dGTP, dCTP) to the 3' end of the DNA primer.
  • deoxynucleotides dATP, dTTP. dGTP, dCTP
  • the capture oligonucleotide was immobilized on the surface of the chip by a dip-coating process.
  • the chips were cleaned.
  • a chip fabricator coated the active surface of the chips with a thin polymer layer to protect it during wafer dicing.
  • the polymer layer was removed by immersing chips in acetone for 5 minutes, followed by isopropanoi for 5 minutes and a second isopropanoi rinse for an additional minute. Chips were then washed in deionized water three times and dried in a vacuum desiccator.
  • RNA standard kanamycin control RNA
  • the capture oligonucleotide primer was complementary to a 24 base sequence near the 3' end of the RNA standard. This sequence is located 971 bases downstream from the 5' end of the RNA standard and contains 224 guanines.
  • the cytosine deoxynucleotides were labeled with a fluorescent dye (Cy5.5). Roche's Transcriptor Reverse Transcriptase was used to extend the primer.
  • the primer extension reaction was carried out at 48° C, which yielded reduced non-specific hybridization. Results from primer extension of 1 pM kanamycin RNA are shown in FIG. 20.
  • the extension rate for the control is slightly negative (-9.7 mV/min), which is probably due to instrument noise.
  • the extension rate of 1 pM kanamycin RNA is 128 mV/min, significantly higher than that of the control.
  • the signal increases linearly over the 1 0.5-minute reaction period with no sign of saturation.
  • the initial signal level of the 1 pM RNA reaction was somewhat lower than that of the control run, and potentially due to a temperature difference ( 1 -2° C.) between the two runs.
  • Quantitative real-time PCR detection of transcripts can be achieved using the detection system of the invention, using a PCR assay protocol modified from that described by Kreuzer et al. (supra).
  • the system includes a substrate chip with excitation waveguides, collection waveguides and intersection regions. The intersection regions where the excitation waveguides and collection waveguides cross include optical sensing sites with sensing wells for conducting quantitative real-time PCR.
  • the detection system further includes a scanning light source which is coupled to and in optical communication with the excitation waveguides of the substrate at some point along its scanning path.
  • the system further includes an optical detector.
  • RNA transcripts of interest e.g., blood from a subject
  • samples suspected of containing RNA transcripts of interest are treated to obtain a source of total RNA which is subsequently reverse transcribed using reverse transcriptase into cDNA using techniques well known in the art.
  • cDNA samples are delivered to sensing sites on the substrate of the system. As desired, suitable controls and different dilutions of a particular cDNA sample can be delivered to different sensing wells.
  • primers/probes specific for one or more bcr/abl breakpoint cluster region.
  • Such primers/probes are well known in the art (e.g., see Kreuzer et al. supra) and can be labeled with 6- carboxy-fluorescein phosphoramidite at the 5' end, and as a quencher, 5-carboxy-tetramethyl-rhodamine can be incorporated further along the primer/probe sequence.
  • a 1 0- ⁇ PCR reaction mix contains 1 ⁇ of 10x PCR buffer, 4.5 mM gC12, 0.8 ni dNTP, 0.5 ⁇ each primer.
  • I ⁇ probe 0.2 units of a temperature-release Taq DNA polymerase (Platinum® Pfx DNA Polymerase; Invitrogen, Corp.), and 20 ng of sample cDNA.
  • PCR ampl ification is started with a 5-min denaturation step at 94° C, followed by 45 cycles of denaturation at 94° C. for 30 s and
  • PCR ampl ification proceeds excitation light at a wavelength of about 658 nm is generated by the scanning light source and is directed lo each sensing well by the excitation waveguides.
  • the cDNA samples include a transcript for bcr/abl fusion transcripts
  • annealing of primers/probes to the cDNA should occur.
  • Subsequent hydrolysis of the probe by polymerase and unquenching of the fluorescein reporter results in fluorescence in the sensing well.
  • the amount of unquenched fluorescein increases in relation to the amount of bcr/abl fusion transcript cDNA in the reaction.
  • fluorescence in the sensing wells is detectable by the optical detector of the system.
  • detection of bcr/abl fusion transcripts can be measured in realtime and based on appropriate controls and analysis the amount of bcr/abl fusion transcripts in a sample can be quantified.
  • Fluorescent immunoassay-based detection of HI V+ status of multiple subjects can be achieved using a detection system such as the example illustrated in FIG. 7A.
  • the system includes a substrate chip with excitation waveguides, collection waveguides and intersection regions. The intersection regions where the excitation waveguides and collection waveguides cross include optical sensing sites with sensing wells for conducting fluorescent immunoassays.
  • the detection system further includes a scanning light source which is coupled to and in optical communication with the substrate at some point along its scanning path and the excitation waveguides.
  • the system further includes an optical detector.
  • a partially purified antigen for example, inactivated HIV protein p29 antigen, is pre- coated onto the sensing wells of the optical sensing sites.
  • subject serums which may contain antibodies to HIV p29 are delivered to separate sensing sites. It is also envisioned that samples from multiple patients are pooled and that each pool is delivered to separate sensing sites. If a subject is H I V+, then their serum may contain antibodies to HIV protein p29, and those antibodies will bind to the H I V p29 antigens on the sensing sites.
  • anti-human immunoglobul in coupled to a fluorescent dye (fluorescein) is added to the sensing sites.
  • This secondary antibody binds to human antibodies in the sensing sites (i.e., the anti-p29 antibodies).
  • an excitation l ight at a wavelength of about 658 ran is generated by the scanning light source and is directed to each sensing well by the excitation waveguides. If the secondary antibody is present the coupled fluorescein will fluoresce in the presence of the excitation light in the well.
  • fluorescence in the sensing wells is detectable by the optical detector of the system.
  • Signal received by the optical detectors can be interpreted to determine i f a given subject, or a sample pooled from multiple subjects, has antibodies to H IV p29.
  • Suitable controls can be used to validate the results of the assay.
  • the presence or absence of HIV p29 can be measured in a sample and H 1V+ or HIV(-) status of multiple subjects or pools can be determined.
  • a two-site sandwich immunoassay can be employed in assays using the detection system of the invention (e.g., the systems as illustrated in FIG. 7A-7E).
  • Antibodies fulfi ll two different roles in such assays, in which an immobilized antibody captures the analyte, while a soluble, fluorescently labeled antibody detects or "traces" analyte binding.
  • capture and tracer antibodies must bind to different sites on the analyte molecule.
  • a single antibody specifically for the repetitive epitope
  • Smaller analytes e.g. proteins and polysaccharides
  • expressing multiple, unique epitopes require two different antibodies, each specific for a unique epitope.
  • Three two-site sandwich immunoassay tests are envisioned: 1 ) serial testing of optical sensing sites; 2) low complexity parallel testing of optical sensing sites; and 3) sensitivity testing.
  • a small volume (1 -5 ⁇ _.) of sample containing analyte and tracer antibody
  • Binding kinetics at the site are monitored over a 5-min period at room temperature.
  • Translation of optical detection to excitation and collection waveguides in connection with a different optical sensing site is effected and the assay is repeated at the new site. It is envisioned that at least 10 optical sensing sites can be tested using this serial procedure. Such tests can demonstrate sensitivity and intra-assay precision of the system.
  • a substrate of the system that includes a 10x 10 array of excitation waveguides and collection waveguides (see e.g., FIG. 7A)
  • a single excitation waveguide of the substrate is excited, while output signals are monitored from all 10 collection waveguides using a linear detector array.
  • Equal volumes (e.g., 50 ⁇ ) of sample containing analyte and tracer antibody (e.g., 10 n , final concentration) can be pre-mixed and then injected into a sample well of the optical sensing site that contains capture antibody. Binding kinetics are monitored simultaneously in 10 optical sensing sites over a 5-min period at room temperature.
  • This form of testing can demonstrate the parallel assay capabilities of the system, as well as providing more detailed information about intra-assay precision.
  • the sensitivity, precision and linearity of the device can be demonstrated by constructing a standard curve of average reaction rate versus analyte concentration.
  • Device configuration is the same as described above for the second form of testing (i.e., 10 simultaneous assays).
  • Analyte concentration is varied over at least a 1 00-fold range, e.g. 1 0 pM to 1 nM, though the exact range can be adjusted depending on the clinical concentration range of the analyte being examined.
  • a separate substrate chip is used for each concentration to be tested. Six to eight concentrations are examined. Resulting standard curves are typically linear at low concentration, but saturate at higher concentrations.
  • the Herron lab has developed immunoassays for many different analytes including human cardiac troponin 1 (cTnl), chorionic gonadotrophin (hCG), creatine phosphokinase isoform M B (CKM B), myoglobin, ovalbumin (used by the mi l itary as a "simulant " for toxins such as ricin and SEB). ricin. Staphylococcal enterotoxin B (SEB). (See Herron, J. N. et al . (2003). Orientation and Activity of I mmobilized Antibodies. In: Biopoiymers at Interfaces, 2nd Edition (M. Malmsten, ed.), Surfactant Science Series, Vol.
  • the ovalbumin assay of Herron can be used in the first and second immunoassays described above.
  • reagents for this assay are relatively inexpensive and no special handling is required.
  • Detection requirements for cTnl and SEB are the most stringent, and thus immunoassays specific for these analytes are useful for the sensitivity testing immunoassays.
  • SEB SEB as a select agent requiring special handling it may be preferable to use cTnl in sensitivity testing.
  • cTnl can be paired with two other cardiac markers (C MB and myoglobin) for simultaneous immunoassay sensitivity testing.
  • FIG. 21 depicts a real-time detection ofClostridiwn difficile toxin A in bovine serum on a 1 0- channel chip.
  • analyte In the first 5 minutes (300 seconds) no analyte is present and thus the signal is flat. After 5- minutes analyte is added to reach a concentration of 10 pM.
  • the analyte binds to the capture antibody immobilized to the chip surface, and a second labeled antibody binds to the complex in a sandwich assay format, the optical signal in all channels increases. The slope of the increasing signal is proportional to the analyte concentration.
  • FIG. 22 shows a standard curve for Clostridium difficile toxin A as measured on a series of ten 10-channel chips.
  • the optical signal slope increases as a function of the increasing concentration.
  • Ten different concentrations were measured, starting from 0.1 pM and up to 1 ⁇ .
  • the slope concentration relation is linear between 0.3 pM and 1 nM.
  • the limit of detection (LOD) is approximately 0. 1 pM.

Abstract

The invention provides methods and devices for generating optical pulses in one or more waveguides using a spatially scanning light source. A detection system, methods of use thereof and kits for detecting a biologically active analyte molecule are also provided. The system includes a scanning light source, a substrate comprising a plurality of waveguides and a plurality of optical sensing sites in optical communication with one or more waveguide of the substrate, a detector that is coupled to and in optical communication with the substrate, and means for spatially translating a light beam emitted from said scanning light source such that the light beam is coupled to and in optical communication with the waveguides of the substrate at some point along its scanning path. The use of a scanning light source al lows the coupling of light into the waveguides of the substrate in a simple and cost-effective manner.

Description

WAVEGUIDE-BASED DETECTION SYSTEM WITH SCANNING LIGHT SOURCE
CROSS REFERENCE TO RELATED APPLICATIONS
[0001] This application is a continuation-in-part of U.S. Patent Application No. 15/245,062, filed August 23, 2016, which is a continuation of U.S. Patent Application No. 14/194,437, filed February 28, 2014, titled "WAVEGUIDE-BASED DETECTION SYSTEM WITH SCANNING LIGHT SOURCE," now U.S. Patent No. 9,423,397, which is a continuation-in-part of U.S. Patent Appl ication No.
12/769,364, filed April 28, 2010, titled "WAVEGUIDE-BASED DETECTION SYSTEM WITH SCANNING LIGHT SOURCE," now U.S. Patent No. 8,675, 199, which claims benefit under 35 U.S.C. § 1 19(e) of U.S. Patent Provisional Application No. 61 /1 73,771 , filed April 29, 2009, titled
"GENERATION OF OPTICAL PULSES IN A WAVEGUIDE USING A SPATIALLY SCANNING LIGHT SOURCE" and U.S. Patent Provisional Application No. 61 /223,550 fi led July 7, 2009 and titled "WAVEGUIDE-BASED DETECTION WITH SCANNING LIGHT SOURCE," each of which is herein incorporated by reference in its entirety.
[0002] U.S. Patent Appl ication No. 12/769,364 is also a continuation-in-part of U.S. Patent
Appl ication No. 1 1 /683,808, filed March 8, 2007, titled "OPTICAL SCANNING SYSTEM," now U.S. Patent No. 7,951 ,583, which claims benefit under 35 U.S.C. § 1 19(e) of U.S. Patent Provisional Appl ication No. 60/743,458, filed March 10, 2006 and titled "OPTICAL SCANNING SYSTEM," each of which is herein incorporated by reference in its entirety.
[0003] U.S. Patent Application No. 12/769,364 is also a continuation-in-part of U.S. Patent Appl ication No. 12/209,295, filed September 12, 2008, titled "WA VEGUIDE-BASED OPTICAL SCANNING SYSTEMS," now U.S. Patent No. 8,288, 157, which claims benefit under 35 U.S.C. § 1 19(e) of U.S. Patent Provisional Application No. 60/971 ,878, filed September 12, 2007, and titled "WAVEGUI DE-BADED OPTICAL SCANNING SYSTEMS," each of which is herein incorporated by reference in its entirety.
[0004] This application also claims priority to U.S. Provisional Application No. 62/235,977, filed October 1 , 201 5, which is herein incorporated by reference in its entirety.
INCORPORATION BY REFERENCE
[0005] A l l publications and patent applications mentioned in this speci fication are herein incorporated by reference to the same extent as if each individual publication or patent application was speci fically and individual ly indicated to be incorporated by reference.
FIELD
[0006] The present invention relates to methods and devices for generati ng optical pulses in one or more waveguides using a spatially scanning l ight source, and to detection systems, methods of use thereof and kits for detecti ng a biologically active analyte molecule. BACKGROUND
[0007] Biological substance analysis methods based on optical means have risen in popularity in the last couple of decades. Common to all these methods is that chemical interactions between the bio- molecules produce changes that affect some measurable optical property, such as the emission spectrum, absorption spectrum or index of refraction. The changes in the optical properties can occur in the analyte itself or through a mediator such as the surface on which the interaction takes place. These changes are then monitored using a beam of incoming light (usually laser light) which in turn changes the outgoing light spectrum (e.g., fluorescence), intensity (e.g., absorption), or phase (e.g., surface plasmon resonance and any kind of interferometric method).
[0008] While most of these optical bio-analysis methods have found niche applications and markets, one method that became highly popular and influential was microarray optical fluorescence scanning Such optical scanning has enabled running tests on tens of thousands of miniature samples in a relatively short period of time. The major advantages of this method include: a) performance (sensitivity and signal to noise ratio (SNR)); b) speed; and c) miniaturization of the sampled analyte. These parameters define the efficiency and superiority of the method.
[0009] Currently microarray elements are spotted on top of a flat substrate chip usually made of glass, plastic or epoxy. Subsequently, the chip is scanned using confocal scanning systems where the exciting light and the resulting fluorescence light are both shined and collected from above and analyzed using a single photo-multiplier (PMT) detector. This arrangement suffers from several inherent limitations including a very short interaction length between the bio-sample and the light (usually a single mono-layer). This limits the signal strength and thus the SNR. Another limitation is a high background or noise due to the fact that the back-reflected light and the emitted fluorescent light travel in the same direction. A further limitation is high sensitivity to both the planarity and the position of the chip that need to be maintained in focus. Stil l another limitation is slow operation due to the need to have large enough number of ' pixels' (scanned spots) within every sample and long enough integration time. Yet another limitation is the need for a complicated optical and mechanical structure that entails bulky and expensive systems.
[00010] Another optical bio-analysis method is waveguide based bio-sensors. Bio-sensing based on waveguides has been around for a while. These biosensors can be divided into three main categories. The first involve slab waveguide fluorescence excitation with light collection from above or belo the chip. In this arrangement the bio-analyzed spots are located on the surface of a chip that contains a single slab- waveguide. Light is coupled into the waveguide using a lens or a grating that excites the entire chip with all its bio-analyzed spots simultaneously. The fluorescence is collected using an optical imaging system and a charge-coupled device (CCD) detector from above or underneath the chip. One drawback of this kind of system is relatively poor performance due to uniformity of excitation as wel l as col lection of the l ight. This leads to non-repeatable results. Another drawback is high noise levels due to crosstalk between the different spots. A further drawback is that large spots and relatively smal l numbers of elements are required to generate a signal large enough for efficient imaging with the CCD. Yet another drawback is the long integration time to overcome SNR issues. Examples of the above method are described in U.S. Pat. Nos. 5,814,565 ; 6,91 1 ,344 and 6,395,558.
[0001 1] A second waveguide based bio-sensor utilizes an interferometric optical device. In this case, channel waveguides are used together with interferometric devices such as Mach Zehnder interferometers (MZ1) or ring-resonators. These sensitive interferometric devices sense the change in the index of refraction due to binding of the bio-molecules near a waveguide surface. The major problems associated with this type of system include non-specificity due to inability to recognize the exact reason for the index change, which may occur from deposition of other material as wel l as temperature changes. Another problem is a very slow speed in addressing the different elements which disqualifies this method for running large numbers of element arrays. Examples of the above method are described in U.S. Pat. Nos. 5,494,798 4,51 5,430, 5,623,561 and 6,618,536.
[00012] A third waveguide based bio-sensor utilizes surface plasmon resonance (SPR). Here, in one example, a thin gold layer is deposited on top of a glass substrate. The bio-analyzed sample on top of the gold induces changes in the refractive index above the gold layer, thus changing the resonant angle for generating surface plasmons along the gold layer. The plasmon generation is detected as an enhanced peak in the reflected beam. Examples of the SPR method are covered, for example, in U.S. Pat. No. 6,956,651 B2. Other types of optical bio-sensors and array scanners exist such as described in U.S. Pat. No. 6,396,995 B l .
[00013] One aspect common to all of these waveguide based sensors is the need to initially couple light into the waveguide. Since all of these optical waveguides have miniature cross-sections ranging from 100 micrometers down to a fraction of a micrometer, the coupling of light into the waveguide involves specialized optics for focusing the light, fine mechanical alignment for accurately placing the light source relative to the waveguide, and specialized glues to bond all components in place without interfering with the light. This process adds in most of these cases a considerable cost and complexity to the entire system.
[00014] In a large number of these optical waveguide applications, the l ight travels in the waveguide in the form of short pulses. These pulses can be as short as a pico ( 1 0~12) second and as long as a few milli ( 10-"1) seconds. Moreover, these pulses can be all of the same wavelength or can be a combination of many different wavelengths. These pulses are generated by modulating one or more light sources which were initial ly coupled to the optical waveguide. If pulses at more than one wavelength are required, a combiner e.g., an Arrayed Waveguide Grating (AWG) must be added to the system.
[00015] In various applications, for example, biological analysis or detection systems, the optical waveguide may be part of a low-cost, consumable chip. Waveguide-based optical detection systems are disclosed, for example, in U .S. Patent Nos. 7,951 ,583 B2, 8, 1 87.866 B2 and 8.288, 1 57 B2. each of which are hereby incorporated in their entirety by reference. In a system making use of such consumable chips, light needs to be coupled time after time to new chips. In such cases the cost and complexity of the l ight coupl i ng technologies known in the art (see, for example. U.S. Pat. Nos. 4.88 1 .789. 5.734.768. 5.600.744. 5,581 ,646, 5.444.805, 5,21 7.568, 5, 1 21 ,457, 5,077,878. 4,744,623 and 4,478,485) are intolerable. [00016] In addition, the optical signal generated on the sensing chip in all these applications is the result of one or more optical phenomena such as fluorescence, luminescence, absorption, scattering and optical phase change. Changes detected in the output light are indicative of the presence of the targets being detected. Nevertheless, the output light may also be affected by additional factors (e.g. temperature and aging) which may skew the results compromising the overall accuracy and precision of the detection system. Finding a method for normalizing the detected signal to overcome the effect of these uncontrol led factors is critical for maintaining the performance of the detection system.
SUMMARY OF THE DISCLOSURE
[00017] The present invention provides methods and devices which allow the coupling of light into one or more optical waveguides in a simple and cost-effective manner. The invention further provides methods and devices for simple coupling of multi-wavelength trains of pulses into an optical waveguide. The invention further provides methods and devices for coupling correlated trains of pulses of one or more wavelengths into multiple optical waveguides.
[00018] In one aspect, the invention provides a method for generating an optical pulse in an optical waveguide, comprising providing an optical waveguide having an internal portion configured to carry an optical signal and a first end face in contact with said internal portion; providing a light beam; and spatially translating the light beam relative to the optical waveguide effective that the light beam transiently contacts the first end face of the optical waveguide, whereby an optical pulse is generated in the waveguide.
[0001 ] In a further embodiment, the invention provides a method for generating an optical pulse in an optical waveguide wherein the light beam has an optical mode, wherein the optical waveguide has an optical mode, and wherein while the light beam transiently contacts the optical waveguide, the light beam optical mode transiently overlaps the optical mode of the waveguide effective that l ight from the light beam passes into and within the optical waveguide.
[00020] In embodiments of the invention, the method further comprises providing a light source capable of emitting a light beam. In some embodiments, the light source is a laser. In some embodiments, the l ight source is a l ight emitting diode (LED). In some embodiments, the l ight beam is reflected prior to contacting the optical waveguide. In some embodiments, the l ight beam is refracted prior to contacting the optical waveguide.
[00021 ] In embodiments of the invention, the l ight source is movable, and spatial translation of the l ight beam emitted from the light source is effected by movement of the light source. In various embodiments, the movement of the l ight source is rotational, vertical, horizontal, transverse or longitudinal. In other embodiments of the invention, the waveguide is movable, and spatial translation of the l ight beam emitted from the l ight source is effected by movement of the waveguide. In various embodiments, the movement of the waveguide is rotational, vertical, horizontal, transverse or longitudi nal . [00022] In embodiments of the invention, the light source is operably connected to an actuator. In other embodiments, the waveguide is operably connected to an actuator. The movement of the actuator may be effected by electrical power, thermal power, magnetic power or even mechanical power (i.e., manually). In various embodiments, the actuator is a piezoelectric based motor, a step motor, an electrical motor, a magnetic actuator, a "memory metal" actuator, a solenoid, or a hydraulic actuator.
[00023] In one embodiment, a light source is mounted on a piezoelectric bending actuator. Applying power to the actuator causes the actuator to move up and down, thereby scanning a line in space. The optical modes of any optical waveguide along this line wil l at some point overlap some of the optical modes of the light source and a pulse of light will be injected into the optical waveguide.
[00024] In a further embodiment, multiple light sources are mounted on a piezoelectric bending actuator. Every time the actuator scans its path, several optical pulses are injected into the optical waveguide. In some embodiments, the multiple light sources each emit light beams having a different wavelength. Every time the actuator scans its path, several optical pulses are injected into the optical waveguide, each at a different wavelength.
[00025] In further embodiments, multiple optical waveguides are arranged along the scanning path of the one or more light sources.
[00026] In an embodiment of the invention, a light source is mounted on the outer edge of a rotating disk, disposed effective that a light beam emitted from the light source is directed outwardly from the disk and transiently contacts an optical waveguide as the disk rotates. In further embodiments, multiple light sources are mounted on the rotating disk. In further embodiments, multiple optical waveguides are mounted around the rotating disk with their optical modes facing inward (toward the center of the disk). With every rotation of the disk, the optical mode of each and every light source wi ll overlap once with the optical mode of each and every optical waveguide. Therefore the total number of pulses generated in each optical waveguide will be equal to the total number of light sources mounted on the disk.
[00027] In further embodiments of the invention, the light beams emitted from the l ight source are spatially translated. In various embodiments, the emitted l ight beams are spatially translated by a lens, a prism, a mirror, or a combination thereof. In some embodiments, the mirror is a stirring mirror.
[00028] In one embodiment, a lens placed in front of the light source is translated in space, causing the light beam emitted by the light source to scan through space unti l its optical mode overlaps the optical mode of an optical waveguide.
[00029] In another embodiment, a stirring mirror placed in front of the light source stirs the emitted light beam through space until its optical mode overlaps the optical mode of the optical waveguide.
[00030] In a further aspect, the invention provides an apparatus for optical pulse generation in an optical waveguide, comprising a light source for emitting a l ight beam; an optical waveguide having a first end; and means for spatial ly translating a light beam from said light source such that the optical mode of the light source transiently contacts said the first end of the optical waveguide effective to provide an optical pulse in the waveguide. [00031] In embodiments of the invention the apparatus further comprises one or more additional light sources, and/or one or more additional optical waveguides. In some embodiments, the apparatus comprises multiple light sources each of which emits light beams having a different wavelength.
[00032] In various embodiments, the means for spatially translating the light beam from the light source relative to the optical waveguide comprises a mechanism selected from a rotating disk, a motor, a solenoid, a hydraulic mechanism, a piezo-electric mechanism, and a "memory-metal" mechanism.
[00033] In an embodiment, the light source is mounted on an outer edge of a rotatable disk, wherein said light source is disposed effective that a light beam emitted by said light source is directed outwardly of said rotatable disk, and that said light beam emitted by said light source is directed effective to transiently contact a first end of an optical waveguide as said disk rotates.
[00034] In a further embodiment, the light source is mounted on an actuator effective that a light beam emitted by said light source is directed effective to transiently contact a first end of an optical waveguide as said actuator moves.
[00035] In a further embodiment, the apparatus further comprises a scanning lens mounted on an actuator, wherein said scanning lens is disposed between the light source and the optical waveguide effective that movement of the actuator causes a light beam emitted by said light source to be directed by the lens effective to transiently contact a first end of an optical waveguide.
[00036] In a further embodiment, the apparatus further comprises a stirring mirror wherein said stirring mirror is disposed between the light source and the optical waveguide effective that movement of the stirring mirror causes a light beam emitted by said light source to be directed by the stirring mirror effective to transiently contact a first end of an optical waveguide.
[00037] In a further aspect, the invention provides optical detection systems which utilize any of the above described methods or devices for coupling light into an optical waveguide. The invention further provides the use of such detection systems for applications including but not limited to detection of a biological marker, detection of a chemical or biological warfare agent, detection or diagnosis of a viral or bacterial infectious disease, diagnosis of a genetic disorder or a cancer, detection of a protein-protein, protein-ligand, or protein-smal l molecule interaction, nucleic acid sequencing, and environmental monitoring of air, water, soil and food samples.
[00038] The present invention further provides detection systems and methods of use thereof including a scanning light source, a detector and a substrate comprising a plurality of waveguides and a plurality of optical sensing sites. The light source is spatially translated relative to the substrate such that l ight emitted from the light source is coupled to and in optical communication with the waveguides of the substrate at some point along its scanning path. The use of a scanning light source allows the coupl ing of l ight into the waveguides of the substrate in a simple and cost-effective manner.
[00039] In general, the invention features a detection system and methods of use thereof including a scanning light source, a substrate comprising a plural ity of waveguides and a plurality of optical sensing sites in optical communication with one or more waveguide of the substrate, a detector that is coupled to and i n optical communication with the substrate, and means for spatial ly translating a l ight beam emitted from said scanning l ight source such that the l ight beam is coupled to and in optical communication with one or more waveguides of the substrate at some point along its scanning path.
[00040] In some embodiments of the invention, the scanning light source is a chip comprising light generating elements. In further embodiments, the scanning light source chip further comprises waveguides. In alternative embodiments, the scanning l ight source is a chip that is further coupled to and in optical communication with an external light source. In some embodiments, the external light source is coupled to the scanning light source chip by optical fibers. In some embodiments, the scanning light source chip further includes waveguides.
[00041] In some embodiments, the substrate comprises a plurality of substantial ly parallel excitation waveguides, and a plurality of substantially parallel collection waveguides, the excitation waveguides and collection waveguides crossing to form a two-dimensional array of intersection regions where an excitation waveguide and a collection waveguide cross and provide optical communication with the intersection region at each crossing; and a plurality of optica! sensing sites each in optical communication with an intersection region. The system further comprises a scanning light source that is at some point along its scanning path coupled to and in optical communication with one or more of the excitation waveguides at a first edge of the substrate, and a detector that is coupled to and in optical communication with one or more of the collection waveguides at a second edge of the substrate. In some embodiments, two or more detectors are coupled to and in optical communication with one or more collection waveguides at various edges of the substrate.
[00042] In other embodiments, the substrate comprises a plurality of substantially parallel waveguides, and a plural ity of optical sensing sites each in optical communication with a waveguide. The system further comprises a scanning light source that is at some point along its scanning path coupled to and in optical communication with one or more of the waveguides at a first edge of the substrate, and a detector that is coupled to and in optical communication with said waveguides at the same or an opposite edge of the substrate. In some embodiments, the substrate comprises a plurality of in-coupl ing waveguides and a plurality of out-coupling waveguides, and a plurality of optical sensing sites each in optical communication with an in-coupling and an out-coupl ing waveguide. The system further comprises a scanning light source that is at some point along its scanning path coupled to and in optical communication with one or more of the in-coupling waveguides at a first edge of the substrate, and a detector that is coupled to and in optical communication with one or more of the outcoupling waveguides of the substrate.
[00043] In some embodiments, the scanning light source further comprises a detector, wherein at the point at which the light source is coupled to and in optical communication with one or more of the plurality of waveguides in optical communication with the optical sensing site, the detector is also coupled to and in optical communication with said one or more waveguides. In some embodiments, the substrate comprises a plurality of in-coupl ing waveguides and a pl ural ity of out-coupl ing waveguides, wherein at the point at which the l ight source is coupled to and in optical commun ication with one or more of the in-coupling waveguides, the detector is coupled to and in optical communication with one or more of the out-coupling waveguides.
[00044] In some embodiments of the invention, the scanning light source is a chip comprising light generating elements and detector elements. In further embodiments, the scanning light source chip further comprises in-coupling and out-coupling waveguides. In further embodiments, the scanning light source chip further comprises at least one combiner. In alternative embodiments, the scanning light source is a chip that is further coupled to and in optical communication with an external light source and an external detector source. In some embodiments, the external light source is coupled to the scanning light source chip by optical fibers. In some embodiments, the scanni ng l ight source chip further includes incoupling and outcoupling waveguides. In further embodiments, the scanning l ight source chip further comprises at least one combiner.
[00045] In some embodiments of the invention, the optical sensing site further comprises a sensor configured to transduce a first light wave generated by the l ight source in a waveguide, resulting in a second light wave in a different waveguide, the second l ight wave being detectable by the detector. In other embodiments, the optical sensing site further comprises a sensor configured to transduce a first light wave generated by the light source in a waveguide, resulting in a second light wave in the same waveguide, the second light wave being detectable by the detector.
[00046] In some embodiments, the light source elements can provide variable wavelengths of light. In some embodiments, the light source is a broad-band source. In other embodiments, the light source is a tunable source. In various embodiments, the l ight source elements may be light emitting diodes (LEDs) or laser diodes (LDs). In various embodiments, the detector elements of the detector or of the scanning light source chip may be PIN diodes, avalanche photo-diodes, or a group of pixels which are part of a charge coupled device (CCD) array. In some embodiments, the detector is a silicon photodiode array.
[00047] In embodiments of the invention, the scanning l ight source is movable, and spatial translation of the light beam emitted from the l ight source is effected by movement of the scanning light source. In further embodiments, spatial translation of the light beam emitted from the l ight source is effected by movement of a component of the scanning light source, such as one or more mirrors, lenses, or prisms. In alternative embodiments, the substrate is movable, and, and spatial translation of the light beam emitted from the light source is effected by movement of the substrate. In various embodiments, the means for spatially translating the light beam from the light source relative to optical waveguides of the substrate comprises a mechanism selected from a rotating disk, a motor, a solenoid, a hydraul ic mechanism, a piezoelectric mechanism, and a memory-metal mechanism. In embodiments of the invention, the scanning l ight source is operably connected to an actuator. The movement of the actuator may be effected by electrical power, thermal power, magnetic power or even mechanical power (i.e.. manual ly). In various embodiments, the actuator is a piezoelectric based motor, a step motor, an electrical motor, a magnetic actuator, a memory metal actuator, a solenoid, or a hydraul ic actuator. In some embodiments, the actuator is a piezoelectric bending actuator. [00048] In general, in yet another aspect, the invention provides a detection method comprising delivering a sample suspected of containing a biologically active analyte molecule to be detected to an optical sensing site on the substrate of a detection system, and spatial ly translating a scanning light source to a point at which the light source is coupled to and in optical communication with one or more of a plurality of waveguides in optical communication with the optical sensing site, thereby generating a first light wave within said waveguide, wherein the first light wave is transducable by a sensor associated with the optical sensing site to a second l ight wave. Furthermore, the method comprises detecting a measurable change in the second light wave using a detector in optical communication with the substrate, wherein a measurable change in the second l ight wave occurs when the sensor interacts with the biologically active analyte molecule.
[00049] In some embodiments, the substrate comprises a plurality of substantially parallel excitation waveguides in optical communication with the optical sensing site, wherein the first light wave is transducable by a sensor associated with the optical sensing site to a second light wave carried in one or more of a plurality of substantially parallel collection waveguides in optical communication with the optical sensing site and crossing the excitation waveguides; and wherein a measurable change in the second light wave is detected using a detector in optical communication with the collection waveguides, wherein a measurable change in the second light wave occurs when the sensor interacts with the biologically active analyte molecule.
[00050] In other embodiments the substrate comprises a plurality of in-coupling and out-coupling waveguides in optical communication with the optical sensing site, and at the point at which the light source is coupled to and in optical communication with one or more of a plurality of in-coupling waveguides in optical communication with the optical sensing site, the detector is coupled to and in optical communication with one or more out-coupling waveguides.
[00051] In other embodiments, the scanning l ight source further comprises a detector, and at the point at which the l ight source is coupled to and in optical communication with one or more waveguides in optical communication with the optical sensing site, the detector is also coupled to and in optical communication with said one or more waveguides. In further embodiments the substrate comprises a plurality of in-coupling and out-coupling waveguides in optical communication with the optical sensing site, and the scanning light source further comprises a detector; and at the point at which the light source is coupled to and in optical communication with one or more of a pl urality of in-coupl ing waveguides in optical communication with the optical sensing site, the detector is coupled to and in optical communication with one or more out-coupl ing waveguides.
[00052] In various embodiments of the methods of the invention, the biologically active analyte is selected from the group consisting of a nucleic acid, a protein, an antigen, an antibody, a microorganism, a gas, a chemical agent and a pollutant. In some embodiments, detecting a measurable change in the second light wave provides a diagnostic result.
[00053] In some embodiments of the invention, a SNP is detected in the biological ly active analyte. In other embodiments of the invention, expression of a gene is detected upon detection of the biologically active analyte. In some embodiments, the method further comprises conducting a real-time PCR reaction at the optical sensing site.
[00054] In some embodiments of the invention, the sensor is adapted to support an immunoassay wherein the sensor interacting with the biologically active analyte comprises an outcome of an immunoassay. In further embodiments, the immunoassay supported is an enzyme-linked immunosorbent assay (ELISA). In further embodiments, the immunoassay supported is a fluorescent immunoassay.
[00055] The invention further provides the use of the detection systems for applications including but not limited to detection of biomarkers, detection of a chemical or biological warfare agent, detection or diagnosis of viral and bacterial infectious diseases, diagnosis of a genetic disorder or a cancer, detection of a protein-protein, protein-ligand, or protein-small molecule interaction, nucleic acid sequencing, and environmental monitoring of air, water, soi l and food samples.
[00056] In some embodiments, a detection system for detecting a biologically active analyte molecule is provided. The system includes a substrate comprising one or more excitation waveguides, a plurality of collection waveguides, the one or more excitation waveguides and the plurality of collection waveguides crossing to form an array of intersection regions where an excitation waveguide and a collection waveguide cross and provide optical communication with the intersection region at each crossing, and a plurality of optical sensing sites each in optical communication with an intersection region; a scanning light source, wherein the scanning l ight source is at some point along its scanning path in optical communication with at least one of the one or more excitation waveguides; a detector that is in optical communication with one or more of the collection waveguides; and an actuator for spatially translating a light beam emitted from the scanning l ight source relative to the substrate such that the light beam is coupled to and in optical communication with at least one of the one or more excitation waveguides of the substrate at some point along its scanning path.
[00057] In some embodiments, the one or more excitation waveguides is a single excitation waveguide.
[00058] In some embodiments, both the one or more excitation waveguides and the plurality of collection waveguides exit the substrate at a first edge of the substrate.
[00059] In some embodiments, the scanning l ight source is at some point along its scanning path in optical communication with one or more of the excitation waveguides at the first edge of the substrate; and the detector is in optical communication with one or more of the collection waveguides at the first edge of the substrate.
[00060] In some embodiments, the one or more excitation waveguides are curved about 90 degrees.
[00061 ] In some embodiments, a detection method is provided. The method includes delivering a sample suspected of containing a biological ly active analyte molecule to be detected to an optical sensing site on a substrate of a detection system, the substrate comprising one or more excitation waveguides and a plurality of col lection waveguides; spatially translating a scanning l ight source to a point at which the l ight source is in optical communication with at least one of the one or more excitation waveguides, wherein at least one of the one or more excitation waveguides is in optical communication with the optical sensing site, thereby generating a first light wave within said at least one of the one or more excitation waveguides, wherein the first light wave is transducable by a sensor associated with the optical sensing site to a second light wave carried in one or more of the plurality of collection waveguides in optical communication with the optical sensing site and crossing the one or more excitation waveguides; and detecting a measurable change in the second light wave using a detector in optical communication with one or more of the plurality collection waveguides, wherein a measurable change in the second light wave occurs when the sensor interacts with the biologically active analyte molecule.
[00062] In some embodiments, the scanning l ight source further comprises a detector, and wherein at the point at which the light source is coupled to and in optical communication with one or more waveguides in optica! communication with the optical sensing site, the detector is also coupled to and in optical communication with said one or more waveguides.
[00063] In some embodiments, the biologically active analyte is selected from the group consisting of a nucleic acid, a protein, an antigen, an antibody, a microorganism, a gas, a chemical agent and a pollutant.
[00064] In some embodiments, the sensor is adapted to support an immunoassay and wherein the sensor interacting with the biologically active analyte comprises an outcome of an immunoassay.
[00065] In some embodiments, the one or more excitation waveguides is a single excitation waveguide.
[00066] In some embodiments, both the one or more excitation waveguides and the plurality of collection waveguides exit the substrate at a first edge of the substrate.
[00067] In some embodiments, the scanning light source is at some point along its scanning path in optical communication with one or more of the excitation waveguides at the first edge of the substrate; and the detector is in optical communication with one or more of the collection waveguides at the first edge of the substrate.
[00068] In some embodiments, the one or more excitation waveguides are curved about 90 degrees.
[00069] The present invention relates to methods and systems of signal normalization, and more particularly to methods and systems of normal izing a signal generated in a waveguide based sensing chip used in bio-sensing applications.
[00070] In some embodiments, a method of measuring a concentration of an analyte using a waveguide chip is provided. The method can include adding a known amount of one or more normalizing moieties to a sample; adding a first detection molecule labelled with a first fluorescent tag to the sample, the first detection molecule specific to the analyte; adding one or more secondary detection molecules, each secondary detection molecule label led with a distinct secondary fluorescent tag, to the sample, the one or more secondary detection molecules specific to the one or more normalizing moieties, wherein the first fluorescent tag and the one or more secondary fluorescent tags emit at different fl uorescence wavelengths: introducing the sample to an array of sensing sites on the waveguide chip, each sensing site of the array having a first capture molecule speci fic to the analyte and one or more secondary capture molecules specific to the one or more normal izing moieties, wherein the first capture molecule and the one or more secondary capture molecules are both immobil ized in each sensing site of the array at a predetermined ratio; introducing a first light pulse into one or more excitation waveguides on the waveguide chip at a first predetermined interval, the one or more excitation waveguides in optical communication with the array of sensing sites, the first light pulse configured to excite the first fluorescent tag; introducing one or more secondary light pulses into the one or more excitation waveguides on the waveguide chip at one or more secondary predetermined intervals, wherein the first l ight pulse and the one or more secondary light pulses have different wavelengths, the one or more secondary light pulses configured to excite the one or more secondary fluorescent tags; measuring the fluorescence emitted by the first detection molecule and the one or more secondary detection molecules through a plural ity of col lection waveguides in optical communication with the array of sensing sites; and normalizing the measured fluorescence emitted by the first detection molecule using the measured fluorescence emitted by the one or more secondary detection molecules.
[00071] In some embodiments, the step of normalizing the measured fluorescence emitted by the first detection molecule includes dividing the measured fluorescence emitted by the first detection molecule by the measured fluorescence emitted by the one or more secondary detection molecules.
[00072] In some embodiments, the first detection molecule, the one or more secondary detection molecules, the first capture molecule, and the one or more secondary capture molecules are antibodies.
[00073] In some embodiments, the step of normal izing the measured fluorescence emitted by the first detection molecule further includes generating one or more calibration curves from the measured fluorescence emitted by the one or more secondary detection molecules.
[00074] In some embodiments, the one or more normalizing moieties includes at least a first normalizing moiety and a second normalizing moiety, wherein the amount of the first normalizing moiety added to the sample is greater than the amount of the second normalizing moiety added to the sample.
[00075] In some embodiments, the step of normal izing the measured fluorescence emitted by the first detection molecule incl udes dividing the measured fluorescence emitted by the first detection molecule by a slope of the measured fluorescence emitted by the one or more secondary detection molecules.
[00076] In some embodiments, the step of normalizing the measured fluorescence emitted by the first detection molecule includes dividing a slope of the measured fluorescence emitted by the first detection molecule by a slope of the measured fluorescence emitted by the one or more secondary detection molecules.
[00077] In some embodiments, a method of measuring a concentration of an analyte using a waveguide chip is provided. The method can include adding a known amount of a first detection molecule label led with a first fluorescent tag to the sample, the first detection molecule specific to the analyte; introducing the sample to an array of sensing sites on the waveguide chip, each sensing site of the array having a first capture molecule speci fic to the analyte and a second capture molecule specific to the normal izing moiety, wherein the first capture molecule and the second capture molecule are both immobi l ized in each sensing site of the array at a predetermined ratio; introducing a first l ight pulse into one or more excitation waveguides on the waveguide chip at a first predetermined interval, the one or more excitation waveguides in optical communication with the array of sensing sites; measuring the fluorescence emitted by the first detection molecule through a plurality of collection waveguides in optical communication with the array of sensing sites; introducing a known amount of a normalizing moiety with a known amount of a second detection molecule labelled with a second fluorescent tag to the array of sensing sites, the second detection molecule specific to the normalizing moiety; introducing a second light pulse into the one or more excitation waveguides on the waveguide chip at a second predetermined interval, the one or more excitation waveguides in optical communication with the array of sensing sites; measuring the fluorescence emitted by the second detection molecule through the plurality of collection waveguides; and normalizing the measured fluorescence emitted by the first detection molecule using the measured fluorescence emitted by the second detection molecule.
[00078] In some embodiments, the step of normalizing the measured fluorescence emitted by the first detection molecule includes dividing the measured fluorescence emitted by the first detection molecule by the measured fluorescence emitted by the second detection molecule.
[00079] In some embodiments, the step of normalizing the measured fluorescence emitted by the first detection molecule includes dividing the measured fluorescence emitted by the first detection molecule by an average of the measured fluorescence emitted by the second detection molecule.
[00080] In some embodiments, the first predetermined interval is the same as the second
predetermined interval .
[00081] In some embodiments, the first predetermined interval is different than the second predetermined interval.
[00082] In some embodiments, the first light pulse and the second light pulse have the same wavelength.
[00083] In some embodiments, a method of measuring a concentration of an analyte using a waveguide chip is provided. The method can incl ude adding a known amount of a first detection molecule labelled with a first fluorescent tag to the sample, the first detection molecule specific to the analyte; introducing the sample to an array of sensing sites on the waveguide chip, each sensing site of the array having an immobil ized capture molecule specific to the analyte; introducing a first light pulse into one or more excitation waveguides on the waveguide chip at a first predetermined interval, the one or more excitation waveguides in optical communication with the array of sensing sites; measuring the fl uorescence emitted by the first detection molecule through a plurality of collection waveguides in optical communication with the array of sensing sites; introducing a known amount of the analyte with a known amount of a second detection molecule labelled with a second fluorescent tag to the array of sensing sites, the second detection molecule speci fic to the analyte; introducing a second light pulse into the one or more excitation waveguides on the waveguide chip at a second predetermined interval, the one or more excitation waveguides in optical communication with the array of sensing sites; measuring the fluorescence emitted by the second detection molecule through the plurality of col lection waveguides; and normal izing the measured fluorescence emitted by the first detection molecule using the measured fl uorescence emitted by the second detection molecule. |00084| In some embodiments, the step of normal izing the measured fluorescence emitted by the first detection molecule includes dividing the measured fluorescence emitted by the first detection molecule by the measured fluorescence emitted by the second detection molecule.
[00085] In some embodiments, the step of normalizing the measured fluorescence emitted by the first detection molecule includes dividing the measured fluorescence emitted by the first detection molecule by a slope of the measured fluorescence emitted by the second detection molecule.
{00086] In some embodiments, the step of normalizing the measured fluorescence emitted by the first detection molecule includes dividing a slope of the measured fluorescence emitted by the first detection molecule by a slope of the measured fluorescence emitted by the second detection molecule.
[00087] In some embodiments, the first light pulse and the second light pulse have the same wavelength.
BRIEF DESCRIPTION OF THE DRAWINGS
[00088] The novel features of the invention are set forth with particularity in the appended claims. A better understanding of the features and advantages of the present methods and compositions may be obtained by reference to the following detailed description that sets forth illustrative embodiments, in which the principles of our methods, compositions, devices and apparatuses are utilized, and the accompanying drawings of which:
[00089] FIG. 1 is a schematic drawing of an optical waveguide.
[00090] FIG. 2A is a schematic drawing illustrating one possible configuration of a light source for use in the invention.
[00091] FIG. 2B is a graph illustrating a typical light source optical mode.
[00092] FIG. 3A is a schematic drawing of an embodiment of a scanning-coupling system according to the invention.
[00093] FIG. 3B is a schematic drawing of a second embodiment of a scanning-coupling system according to the invention including multiple optical waveguides.
[00094] FIG. 3C is a schematic drawing of a third embodiment of a scanning-coupling system according to the invention including multiple light sources and multiple optical waveguides.
[00095] FIG. 4A is a schematic drawing of a fourth embodiment of a scanning-coupling system according to the invention including a rotating disk.
[00096] FIG. 4B is a schematic drawing of a fifth embodiment of a scanning-coupling system according to the invention including a rotating disk, multiple light sources and multiple optical waveguides.
[00097] FIG. 5 is a schematic drawing of a sixth embodiment of a scanning-coupl ing system according to the invention including a scanning lens.
[00098] FIG. 6 is a schematic drawing of a seventh embodiment of a scanning-coupl ing system according to the invention includi ng a stirring mirror. [00099] FIG. 7A is a schematic of a detection system according to one embodiment of the invention including an active scanning light source chip, a substrate, a detector, and optical sensing sites.
[000100] FIG. 7B is a schematic of a detection system according to another embodiment of the invention including an active scanning l ight source/detector chip, a substrate and optical sensing sites.
[000101 ] FIG. 7C is a schematic of a detection system according to another embodiment of the invention including a light source, fibers, a passive scanning light source chip, a substrate, optical sensing sites and a detector.
[000102] FIG. 7D is a schematic of a detection system according to another embodiment of the invention including a light source, a detector, fibers, a passive light source/detector chip, a substrate and optical sensing sites.
[000103] FIG. 7E is a schematic side view of a detection system according to an embodiment of the invention including a scanning light source/detector chip, a piezoelectric bending actuator and a substrate.
[000104] FIG. 7F is a schematic of another embodiment of a substrate having a single excitation waveguide and a plurality of collection waveguides that exit the same side of the substrate.
[000105] FIG. 7G is a schematic of another embodiment of a substrate having a plurality of excitation waveguides and a pl ural ity of collection waveguides that exit the same side of the substrate.
[000106] FIG. 7H is a schematic of an embodiment of a substrate having excitation waveguides and collection waveguides and a loopback waveguide that exits the same side of the substrate.
[000107] FIG. 8 is a block diagram showing a representative example of a detection system of the invention in a housing as part of a working system.
[000108] FIG. 9A is a schematic of the substrate of the invention according to one embodiment including optical waveguides in conjunction with optical sensing sites and barriers.
[000109] FIG . 9B is a schematic of the substrate of the invention according to another embodiment including optical waveguides and combiners in conjunction with optical sensing sites and barriers.
[0001 10] FIG. 9C is a schematic cross section of the substrate of the invention according to one embodiment including an optical waveguide in conjunction with an optical sensing site.
[0001 1 1 ] FIG. 9D is a schematic of the substrate of the invention according to another embodiment including excitation and collection optical waveguides in conjunction with optical sensing sites and barriers.
[0001 12 ] FIG. 9E is a perspective view of the substrate of the embodiment of the invention shown in FIG. 9D including excitation and collection optical waveguides in conj unction with optical sensing sites.
[0001 13] FIG. 9F is a schematic of two cross sectional views (AA and BB) of the substrate shown in FIGS. 9D and 9E.
[0001 14] FIG . 9G is a schematic of a side view of the substrate of one embodiment of the invention in relation to a thermoelectric cooler. [0001 15] FIG. 9H is a schematic of one embodiment of the substrate of the invention illustrating details of an optical sensing site including a heater and a thermistor.
[000116] FIG. 91 is a schematic of another embodiment of the substrate of the invention illustrating details of an optical sensing site including a heater and a thermistor.
[000117] FIG. 9J is a schematic of one embodiment of the substrate of the invention including reservoirs and micro channels in relation to optical sensing sites.
[0001 18] FIG. 9K is a schematic of another embodiment of the substrate of the invention including reservoirs and micro channels in relation to optical sensing sites.
[000119] FIG. 10A is a schematic of the substrate of the invention including excitation and collection optical waveguides in conjunction with optical sensing sites, barriers and funnels.
[000120] FIG. 10B is a schematic showing an enlarged view of substrate features according to an embodiment as shown in FIG. 10A.
[000121 ] FIG. I OC is a schematic of a cross-sectional view of the substrate according to one embodiment.
[000122] FIG. 1 1 A is a schematic of one embodiment of the substrate of the invention including excitation and collection optical waveguides in conjunction with optical sensing sites, barriers and branches.
[000123] FIG. 1 1 B is a schematic showing an enlarged view of substrate features according to an embodiment as shown in FIG. 1 1 A.
[000124] FIG. 1 1 C is a schematic of a cross-sectional view in a plane (AA) of the substrate according to one embodiment.
|000125] FIG. 1 1 D is a schematic of a cross-sectional view in a plane (BB) of the substrate according to one embodiment.
[000126] FIG. 12A is a schematic of a passive scanning light source chip of the invention according to one embodiment including optical in-coupling waveguides.
[000127] FIG. 12B is a schematic of a passive scanning light source/detector chip of the invention according to one embodiment including optical in-coupling and out-coupling waveguides and optical combiners.
[000128] FIG. 12C is a schematic of a passive scanning light source/detector chip of the invention according to another embodiment including optical in-coupling and out-coupling waveguides.
[000129] FIG. 13A is a schematic of a general substrate including typical layers and waveguides representative of those of the current invention.
1000130] FIG. 1 3 B is a photomicrograph image of waveguides representative of those of the invention and a silica layer.
|000131 ] F IG. 13C is a perspective view of waveguides and associated substrate layers.
[000132] FIG. 14A is a schematic of an active scanning l ight source chip of the invention according to one embodiment including l ight source elements and optical in-coupl ing waveguides. [000133] FIG. 14B is a schematic of an active scanning l ight source/detector chip of the invention according to one embodiment including light source elements, detector elements, optical in-coupling and out-coupl ing waveguides and optical combiners.
[000134] FIG. 15 is a block diagram showing a representative example logic device in communication with an apparatus for use with the detection system of the invention.
[000135] FIG. 16 is a block diagram showing a representative example of a kit.
[000136] FIGS. 17A-D are schematics illustrating a representative manufacturing process for the substrate and waveguides of the invention.
[000137] FIG. 18 is a flow chart showing a representative manufacturing process for the substrate.
[000138] FIG. 19 is a plot of data for fluorescently labeled ovalbumin binding to immobilized antibodies specific for ovalbumin.
[000139] FIG. 20 is a plot of data for incorporation of Cy5.5-labeled cytosines into a DNA molecule during a primer extension reaction as detected using one embodiment of a detection system of the invention.
[000140] FIG. 21 is a graph showing a real-time detection of Clostridium difficile toxin A in bovine serum on a 10-channel chip using a detection system of the invention.
[000141 ] FIG. 22 is a graph showing a standard curve for Clostridium difficile toxin A as measured on a series of ten 10-channel chips using a detection system of the invention.
[000142] FIG. 23 is a flow chart illustrating a procedure for aligning a cartridge in a reader.
[000143] FIG. 24 is a flow chart illustrating a procedure for immobilizing antibodies to a substrate.
[000144] FIGS. 25A-25D illustrate an array of droppers that can dispense droplets of antibodies onto a substrate.
DETAILED DESCRIPTION
[000145] The present invention provides methods and devices for coupling light of one or more wavelengths into one or more optical waveguides in a simple and cost-effective manner. Apparatus, methods, and kits for optical detection, using a detection system including a scanning light source, a detector, a substrate, and a plurality of waveguides and optical sensing sites are also provided. One substrate of the present system includes a plurality of substantially parallel excitation waveguides and a plurality of substantially parallel collection waveguides. The excitation waveguides and collection waveguides cross to form an intersection region and a two-dimensional array. Other substrates of the present system include a plurality of substantially parallel waveguides and a plural ity of sensing sites. The optical sensing sites include a sensor and are in optical communication with one or more waveguides. Detection of a variety of environmental and biological samples can be achieved using the apparatus, methods and kits described herein. The general theoretical principles of lightwave guiding and evanescent field fluorescence excitation apply to the embodiments disclosed herein.
[0()0146| As used herein and in the appended claims, the singular forms "a,"" "an," and ltthe'" include pl ural reference unless the context clearly indicates otherwise. [000147] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood to one of ordinary skill in the art to which the inventions described herein belong. Although any methods, devices, and materials similar or equivalent to those described herein can be used in the practice or testing of the inventions described herein, the preferred methods, devices and materials are now described.
Definitions
[000148] The term "biologically active analyte" when used herein means any substance which can affect any physical or biochemical properties of a biological organism, including but not limited to viruses, bacteria, fungi, plants, animals, and humans. In particular as used herein, biologically active analyte according to the present invention includes without limitation drugs, prodrugs, pharmaceutical agents, drug metabolites, biomarkers such as expressed proteins and cell markers, antibodies, serum proteins, cholesterol, polysaccharides, nucleic acids, biological analytes, genes, proteins, or hormones, or any combination thereof. A biologically active analyte can further include a natural or man-made substance including but not limited to a gas, a chemical agent or a pollutant, or a combination thereof (e.g., from an environmental source). At a molecular level, the biologically active analytes can be polypeptide, glycoprotein, polysaccharide, lipid, nucleic acid, or a combination thereof.
[000149] Of particular interest are biomarkers associated with a particular disease or with a specific disease stage.
[000150] Such biologically active analytes include but are not limited to those associated with autoimmune diseases, obesity, hypertension, diabetes, neuronal and/or muscular degenerative diseases, cardiac diseases, endocrine disorders, and any combinations thereof.
[000151] Also of interest are biomarkers that are present in varying abundance in one or more of the body tissues including heart, liver, prostate, lung, kidney, bone marrow, blood, skin, bladder, brain, muscles, nerves, and selected tissues that are affected by various disease, such as different types of cancer (malignant or non-metastatic), autoimmune diseases, inflammatory or degenerative diseases.
|000152] Also of interest are biologically active analytes that are indicative of a microorganism. Exemplary microorganisms include but are not limited to bacterium, virus, fungus and protozoa.
Biologically active analytes that can be detected by the subject method also include blood-born pathogens selected from a non-limiting group that consists of Staphylococcus epiclermidis, Escherichia
coli, methici llin-resistant Staphylococcus aureus (MSRA), Staphylococcus aureus, Staphylococcus hominis, Enterococcus faecalis, Pseudomonas aeruginosa, Staphylococcus capitis, Staphylococcus warneri, Klebsiella pneumoniae, Haemophilus influnzae, Staphylococcus simulcms, Streptococcus pneumoniae and Candida albicans.
|000153] Biologically active analytes that can be detected by the subject device and methods also encompass a variety of sexually transmitted diseases selected from the fol lowing: gonorrhea (Neisseria gorrhoeae), syphil is { Treponena pallidum), chlamydia (Chlamydia iracomi/is), nongonococcal urethritis ( Ureaplasma urealyticum), yeast infection (Candida albicans), chancroid (Haemophilus ducreyi), trichomoniasis (Trichomonas vaginalis), genital herpes (HSV type I and II), H IV I, HI V II and hepatitis A, B, C, G, as well as hepatitis caused by TTV.
|000154] Additional biologically active analytes that can be detected by the subject apparatus and methods encompass a diversity of respiratory pathogens including but not limited to Pseudomonas aeruginosa, methici llin-resistant Staphylococcus aureus(MSRA), Klebsiella pneumoniae, Haemophilis influenzae, Staphylococcus aureus, Stenotrophomonas maltophilia, Haemophilis parainfluenzae, Escherichia coli, Enterococcus faecalis, Serratia marcescens, Haemophilis par ahaemolyticus,
Enterococcus cloacae, Candida albicans, Moraxiella catarrhalis, Streptococcus pneumoniae, Citrobacter freiindii, Enterococcus faecium, Klebsiella oxytoca, Pseudomonas fluorsecens, Neisseria meningitidis, Streptococcus pyogenes, Pneumocystis carinii, Klebsiella pneumoniae, Legionella pneumophila, Mycoplasma pneumoniae, and Mycobacterium tuberculosis.
[000155] Listed below are additional exemplary markers according to the present invention: Theophylline, CRP, C MB, PSA, Myoglobin, CA 125, Progesterone, TxB2, 6-keto-PGF-l -alpha, and Theophylline, Estradiol, Lutenizing hormone, High sensitivity CRP, Triglycerides, Tryptase, Low density l ipoprotein Cholesterol, High density lipoprotein Cholesterol, Cholesterol, IGFR.
[000156] Exemplary liver markers include without limitation LDH, (LD5), (ALT), Arginase 1 (liver type), Alphafetoprotein (AFP), Alkaline phosphatase, Alanine aminotransferase, Lactate dehydrogenase, and Bilirubin.
[000157] Exemplary kidney markers include without limitation TNFa Receptor, Cystatin C, Lipocalin-type urinary prostaglandin D, synthatase (LPGDS), Hepatocyte growth factor receptor, Polycystin 2, Polycystin 1 , Fibrocystin, Uromodulin, Alanine, aminopeptidase, N-acetyl-B-D- glucosaminidase, Albumin, and Retinol-binding protein (RBP).
[000158] Exemplary heart markers include without limitation Troponin I (Tnl), Troponin T (TnT), CK, CKMB, Myoglobin, Fatty acid binding protein (FABP), CRP, D-dimer, S- 100 protein, BNP, NT- pro BNP, PAPP-A, Myeloperoxidase (MPO), Glycogen phosphorylase isoenzyme BB (GPBB), Thrombin Activatable Fibrinolysis Inhibitor (TAFI), Fibrinogen, Ischemia modified albumin (IMA), Cardiotrophin- 1 , and LC-1 (Myosin Light Chain-I).
]000159] Exemplary pancreas markers include without limitation Amylase, Pancreatitis-Associated protein (PAP-1 ), and Regenerated proteins (REG).
[000160] Exemplary muscle tissue markers include without limitation Myostatin.
[000161] Exemplary blood markers include without limitation Erythopoeitin (EPO).
[000162] Exemplary bone markers include without limitation, Cross-l inked N-telopeptides of bone type I col lagen (NTx), Carboxyterminal cross-linking telopeptide of bone collagen, Lysyl-pyridinol ine (deoxypyridinol ine), Pyridinol ine, Tartrate-resistant acid phosphatase, Procollagen type I C propeptide,
Procollagen type I N propeptide, Osteocalcin (bone gla-protein), Alkal ine phosphatase, Cathepsin K,
CO P (Cartilage Oligomeric Matrix Protein), Osteocrin, Osteoprotegerin (OPG), RA L, sRANK, TRAP 5 (TRACP 5), Osteoblast Specific Factor 1 (OSF-1 , Pleiotrophin), Soluble cell adhesion molecules, sTfR, sCD4, sCD8, sCD44, and Osteoblast Specific Factor 2 (OSF-2, Periostin).
[000163] In some embodiments markers according to the present invention are disease specific. Exemplary cancer markers include without limitation PSA (total prostate specific antigen), Creatinine, Prostatic acid phosphatase, PSA complexes, Prostrate-specific gene- 1 , CA 12-5, Carcinoembryonic
Antigen (CEA), Alpha feto protein (AFP), hCG (Human chorionic gonadotropin), Inhibin, CAA Ovarian C 1 824, CA 27.29, CA 15-3, CAA Breast C 1924, Her-2, Pancreatic, CA 19-9, Carcinoembryonic Antigen, CAA pancreatic, Neuron-specific enolase, Angiostatin. DcR3 (Soluble decoy receptor 3), Endostatin, Ep- CAM ( K- 1 ), Free Immunoglobulin Light Chain Kappa, Free Immunoglobulin Light Chain Lambda, Herstatin, Chromogranin A, Adrenomedullin, Integrin, Epidermal growth factor receptor, Epidermal growth factor receptor-Tyrosine kinase, Pro-adrenomedullin N-terminal 20 peptide, Vascular endothelial growth factor, Vascular endothelial growth factor receptor, Stem cell factor receptor, c-kit/KDR, KDR, and Midkine.
[000164] Exemplary infectious disease markers include without limitation Viremia, Bacteremia, Sepsis, PMN Elastase, PMN elastase/al -Pl complex, Surfactant Protein D (SP-D), HBVc antigen, HBVs antigen, Anti-HBVc, Anti-HIV, T-suppressor cell antigen, T-cell antigen ratio, T-helper cell antigen, Anti-HCV, Pyrogens, p24 antigen and Muramyldipeptide.
[000165] Exemplary diabetes markers include without limitation C-Peptide, Hemoglobin Ale,
Glycated albumin, Advanced glycosylation end products (AGEs), 1 ,5-anhydroglucitol, Gastric Inhibitory Polypeptide, Glucose, Hemoglobin, ANGPTL3 and ANGPTL 4.
[000166] Exemplary inflammation markers include without limitation Rheumatoid factor (RF),
Antinuclear Antibody (ANA), C-reactive protein (CRP) and Clara Cell Protein (Uteroglobin).
[000167] Exemplary allergy markers include without limitation Total IgE and Specific IgE.
[000168| Exemplary autism markers include without l imitation Ceruloplasmin, Metalothioneine, Zinc, Copper, B6, B 12, Glutathione, Alkaline phosphatase, and Activation of apo-alkaline phosphatase.
[000169] Exemplary coagulation disorders markers include without limitation b-Thromboglobulin,
Platelet factor 4 and Von Willebrand factor.
[000170] In some embodiments a marker may be therapy specific. COX inhibitors include without limitation TxB2 (Cox- 1 ), 6-keto-PGF-l -alpha (Cox 2) and 1 1 -Dehydro-TxB- l a (Cox- 1 ).
[000171 ] Other markers of the present incl ude without l imitation Leptin, Leptin receptor,
Procalcitonin, Brain S I 00 protein, Substance P and 8-Iso-PGF-2a.
[000172] Exemplary geriatric markers include without limitation, Neuron-specific enolase, GFAP and S 100B.
[000173 | Exemplary markers of nutritional status incl ude without limitation Prealbumin, Albumin, Retinol-binding protein (RBP), Transferrin, Acylation-Stimulating Protein (ASP), Adiponectin, Agouti- Related Protein (AgRP), Angiopoietin-like Protein 4 (ANGPTL4, FIAF), C-peptide, AFABP (Adipocyte Fatty Acid Binding Protein, FAB P4). Acylation-Stimulating Protein ( ASP). EFA BP ( Epidermal Fatty Acid Binding Protein, FABP5), Glicentin, Gl ucagon. G lucagon-Like Peptide- 1 . Gl ucagon-Like Peptide-2, Ghrelin, Insulin, Leptin, Leptin Receptor, PYY, RELMs, Resistin, and sTfR (soluble Transferrin Receptor).
[000174] Exemplary markers of lipid metabolism include without limitation Apo-lipoproteins (several), Apo-A l , Apo-B, Apo-C-Cll, Apo-D and Apo-E.
[000175] Exemplary coagulation status markers include without limitation Factor 1 : Fibrinogen, Factor II : Prothrombin, Factor II I : Tissue factor, Factor IV: Calcium, Factor V: Proaccelerin, Factor VI, Factor VII: Proconvertin, Factor VIII :, Anti-hemolytic factor, Factor IX : Christmas factor, Factor X: Stuart-Prower factor, Factor XI : Plasma thromboplastin antecedent, Factor XI I : Hageman factor, Factor XI I I: Fibrin-stabilizing factor, Prekallikrein, High-molecular-weight kininogen, Protein C, Protein S, D- dimer, Tissue plasminogen activator, Plasminogen, a2-Antiplasmin and Plasminogen activator inhibitor 1 (PAI 1 ).
[000176] Exemplary monoclonal antibody markers include those for EGFR, ErbB2, and IGF 1 R.
[000177] Exemplary tyrosine kinase inhibitor markers incl ude without limitation Abl, Kit, PDGFR, Src, ErbB2, ErbB 4, EGFR, EphB, VEGFRl -4, PDGFRb, FLt3, FGFR, PKC, Met, Tie2, RAF, and TrkA.
[000178] Exemplary serine/threonine kinase inhibitor markers include without limitation AKT, Aurora A/B/B, CDK, CDK (pan), CD l -2, VEGFR2, PDGFRb, CD 4/6, MEK 1 -2, mTOR, and PKC- beta.
[000179] GPCR target markers include without limitation Histamine Receptors, Serotonin Receptors, Angiotensin Receptors, Adrenoreceptors, Muscarinic Acetylcholine Receptors, GnRFl Receptors, Dopamine Receptors, Prostaglandin Receptors, and ADP Receptors.
[000180] For the purposes of this invention, a "therapeutic agent" is intended to include any substances that have therapeutic utility and/or potential. Such substances include but are not limited to biological or chemical compounds such as simple or complex organic or inorganic molecules, peptides, proteins (e.g. antibodies) or polynucleotides (e.g. anti-sense). A vast array of compounds can be synthesized, for example, polymers, such as polypeptides and polynucleotides, and synthetic organic compounds based on various core structures, and these are also included in the term "therapeutic agent". In addition, various natural sources can provide compounds for screening, such as plant or animal extracts, and the like. It should be understood, although not always explicitly stated, that the agent is used alone or in combination with another agent, having the same or different biological activity as the agents identified by the inventive screen. The agents and methods also are intended to be combined with other therapies.
[000181 ] Pharmacodynamic (PD) parameters according to the present invention include without l imitation physical parameters such as temperature, heart rate/pulse, blood pressure, and respiratory rate, and biomarkers such as proteins, cells, and cell markers. Biomarkers could be indicative of disease or could be a result of the action of a drug. Pharmacokinetic (PK) parameters according to the present invention include without l imitation drug and drug metabolite concentration. Identifying and quantifying the PK parameters rapidly from a sample vol ume is extremely desirable for proper safety and efficacy of drugs. If the drug and metabol ite concentrations are outside a desired range and/or unexpected metabol ites are generated due to an unexpected reaction to the drug, immediate action may be necessary to ensure the safety of the patient. Similarly, if any of the PD parameters fall outside the desired range during a treatment regime, immediate action may have to be taken as well.
[000182] In preferred embodiments physical parameter data is stored in or compared to stored profiles of physical parameter data in a bioinformatics system which may be on an external device incorporating pharmacogenomic and pharmacokinetic data i nto its models for the determination of toxicity and dosing. Not only does this generate data for clinical trials years prior to current processes but also enables the elimination of current disparities between apparent efficacy and actual toxicity of drugs through real-time continuous monitoring. During the go/no go decision process in clinical studies, large scale comparative population studies can be conducted with the data stored on the database. This compilation of data and real-time monitoring allows more patients to enter clinical trials in a safe fashion earlier than currently allowed. In another embodiment biomarkers discovered in human tissue studies can be targeted by the detection system for improved accuracy in determining drug pathways and efficacy in cancer studies.
[000183] The term "nucleic acid" when used herein refers to deoxyribonucleotides,
deoxyribonucleosides, ribonucleosides or ribonucleotides and polymers thereof in either single- or double-stranded form. Unless specifically limited, the term encompasses nucleic acids containing known analogues of natural nucleotides which have similar binding properties as the reference nucleic acid and are metabolized in a manner similar to naturally occurring nucleotides. Unless specifically limited otherwise, the term also refers oligonucleotide analogs including PNA (peptidonucleic acid), analogs of DNA used in antisense technology (phosphorothioates, phosphoroamidates, and the like). Unless otherwise indicated, a particular nucleic acid sequence also impl icitly encompasses conservatively modified variants thereof (including but not limited to, degenerate codon substitutions) and
complementary sequences as well as the sequence explicitly indicated. Specifically, degenerate codon substitutions may be achieved by generating sequences in which the third position of one or more selected (or all) codons is substituted with mixed-base and/or deoxyinosine residues (Batzer et al., Nucleic Acid Res. 19:5081 ( 1991 ); Ohtsuka et al., J. Biol. Chem. 260:2605-2608 ( 1 985); and Rossolini et al ., Mol. Cell. Probes 8:91 -98 ( 1994)).
[000184] The term "microorganism'" when used herein refers to bacteria, actinomycetales, cyanobacteria (unicel lular algae), fungi, protozoa, animal cells or plant cells or viruses. Examples of microorganisms include but are not limited to pathogens.
[000185] The terms "polypeptide," "peptide" and "protein" are used interchangeably herein to refer to a polymer of amino acid residues. That is, a description directed to a polypeptide applies equally to a description of a peptide and a description of a protein, and vice versa. The terms apply to naturally occurring amino acid polymers as well as amino acid polymers in which one or more amino acid residues is a non-natural amino acid. As used herein, the terms encompass ami no acid chains of any length, including ful l length proteins (i.e., antigens), wherein the amino acid residues are linked by covalent peptide bonds. In addition, proteins that contain multiple polypeptide chains that associate through covalent and/or non-covalent interactions are also encompassed by "protein," as used herein.
[000186] The term "polymorphism" as used herein refers to the occurrence of two or more genetically determined alternative sequences or alleles in a population. A polymorphic marker or site is the locus at which divergence occurs. Preferred markers have at least two alleles, each occurring at a frequency of greater than 1 %, and more preferably greater than 10% or 20% of a selected population. A polymorphism may comprise one or more base changes, an insertion, a repeat, or a deletion. A polymorphic locus may be as small as one base pair. Polymorphic markers include restriction fragment length polymorphisms, variable number of tandem repeats (VNTR's), hypervariable regions, minisatellites, dinucleotide repeats, trinucleotide repeats, tetranucleotide repeats, simple sequence repeats, and insertion elements such as Alu. The first identified allelic form is arbitrarily designated as the reference form and other allelic forms are designated as alternative or variant alleles. The allelic form occurring most frequently in a selected population is sometimes referred to as the wildtype form. Diploid organisms may be homozygous or heterozygous for al lelic forms. A diallelic polymorphism has two forms. A triallelic polymorphism has three forms.
[000187] A single nucleotide polymorphism (SIMP) occurs at a polymorphic site occupied by a single nucleotide, which is the site of variation between allelic sequences. The site is usually preceded by and followed by highly conserved sequences of the allele (e.g., sequences that vary in less than 1 /100 or 1 /1000 members of the populations).
[000188] A single nucleotide polymorphism usually arises due to substitution of one nucleotide for another at the polymorphic site. A transition is the replacement of one purine by another purine or one pyrimidine by another pyrimidine. A transversion is the replacement of a purine by a pyrimidine or vice versa. Single nucleotide polymorphisms can also arise from a deletion of a nucleotide or an insertion of a nucleotide relative to a reference al lele.
[000189] The term "individual" when used herein is not limited to a human being, but may also include other organisms including but not limited to mammals, plants, bacteria or cells derived from any of the above.
[000190] The term "about" or "approximately" as used herein can mean within 10, 20, or 30 percent.
[000191] Aspects of the invention may include one or more of the fol lowing advantageous features. Dense and accurate integration of optical manipulating elements can be achieved using planar lightwave circuit technology. Appl ications for planar lightwave circuits as described herein include new drug discovery and development, disease research, biomarkers discovery, detection of a chemical or biological warfare agent, environmental monitoring, SNP association studies including toxicology and disease susceptibi l ity, and diagnostics incl uding identifying patients predisposed to diseases and identifying patients with particular drug sensitivity.
[000192] "Optical coupling" from one clement to another (e.g. a light emitter to an opt ical waveguide) occurs whenever there is some overlap between the "optical modes" of the two elements. The "optical mode" of an element, as defined herein, represents the spatial and temporal behavior of the light emitted from or accepted by that element.
[000193] In the current invention one or more light sources or alternatively, their emitted light beams, are spatially translated relative to one or more optical waveguides with the optical modes of the light sources scanning through space to overlap at some arbitrary time and arbitrary place with some of the optical modes of one or more optical waveguides. During that overlap, light is coupled from the overlapping optical mode of a light source to the overlapping optical mode of an optical waveguide, thereby generating a light pulse within the optical waveguide, which pulse has a duration equal to the overlapping time and a wavelength equal to that of the overlapping light source.
[000194] By "spatially translated relative to the optical waveguide" is meant that either the light sources are physically translated through space, or their emitted light beams are translated using optical means (e.g. lenses, prisms, mirrors, etc.) or the optical waveguides are physically translated through space, until the overlap of the optical modes occurs.
[000195] An apparatus which utilizes this method of generating an optical pulse using a spatially scanning light beam is referred to herein as the "scanning-coupling system" of the invention.
[000196] A general optical waveguide 101 is depicted in FIG. 1 . It consists of two main regions, the core region 1 1 1 and the cladding region 1 10. Light is confined and propagates along a waveguide in the core region. This confinement is most commonly achieved by designing the core region to have an index of refraction higher than that of the cladding region. Thus light 'trying to escape' the core region experiences 'total internal reflection' and remains trapped in the core region. Only light having certain properties (e.g., propagation angle) is trapped within the core region. Every optical waveguide has a discrete set of "modes" having those properties and thus able to propagate along the waveguide. These are referred to as the "optical modes" of the waveguide 1 12.
[000197] A representative light source 202 is schematically shown in FIG. 2A. The light source consists of a laser chip 223 and the electrical leads 221 for driving it. Although lasers are the most common source for coupling light into a waveguide, in some cases a Light Emitting Diode (LED) may be used. A lens 222 in the drawing represents a variety of optical components that can be used to manipulate (i.e. collimate, focus, filter, deflect, etc.) the emitted light. Al l these components can be assembled in a compact way on a single or on multiple substrates 224.
[000198] Such a light source is connected to its driving electronics which control the way it operates to generate a continuous wave (CW) of light or pulses. The l ight source generates l ight in one or in several different wavelengths. In some cases, the wavelength of the l ight emitted by the light source can be tuned by changing its temperature or by placing a filter in front of the light source to pick one wavelength out of the emitted spectrum. The light emitted by the light source and passing through the lens (and/or other optical components) has a certain intensity distribution and propagation angles, referred to herein as the light source "optical mode". FIG. 2B shows an example of a l ight source optical mode 21 3, its intensity distribution 21 4 at a given distance (Z). its Y distribution 21 5 and its X distribution 2 1 6. [000199] In embodiments of the present invention, a pulse of light from the light source is coupled to the optical waveguide by moving the one relative to the other through a point where there is some overlap between the optical modes of the two, resulting in the generation of an optical pulse within the optical waveguide. Repeating the same process with the same light source and optical waveguide generates a train of identical light pulses. Repeating the same process with different light sources generates a train of pulses differing in wavelength, and/or duration and/or any other property of the light emitted by the light sources (e.g., intensity, temporal coherence, spatial coherence, or amplitude modulation). Repeating the scanning process with multiple light sources and multiple optical waveguides generates correlated trains of pulses in all or some of the optical waveguides.
[000200] The generated pulse will be a convolution in space of the two optical modes of the light source and the optical waveguide. This convolution is affected by both of the optical modes and their spatial and temporal overlap. The resulting pulse is also affected by the size of the two beams. The scanning speed also affects the generated pulse through the temporal dependence of the moving optical mode. If the waveguide has more than one mode the resulting pulse will be a sum of all overlaps.
[000201] The pulse duration may be more simply calculated by taking the effective beam diameter of the light source and that of the waveguide. The approximate pulse duration is the sum of these two beam diameters divided by the scanning amplitude, multiplied by the scanning period.
[000202] There are many possible means for achieving the relative movement of the one or more light sources or their emitted light beams with respect to the one or more optical waveguides. These means may include mechanisms such as, for example, a rotating disk, a motor, a solenoid, a hydraulic mechanism, a piezo-electric mechanism, and a "memory-metal" mechanism. In addition, the relative motion may be generated through simple manually driven actuators. Either the light sources or the optical waveguides may be physically translated through space. Alternatively, the light sources may be stationary while the light beams emitted from the light sources are translated using optical means (e.g. lenses, prisms, mirrors, etc.). Spatial translation of the optical means may be achieved using any of the above described means.
[000203] Various non-limiting embodiments of the optical coupling methods and devices of the invention are described below.
[000204] In one embodiment of the invention, the light source 302 is mounted on a piezoelectric bending actuator 305 as shown in FIG. 3A. The scanning-coupling system 300 further comprises an optical waveguide 301 . The piezoelectric bending actuator driver (not shown) interfaces to the actuator through electrical wires 306. The driver generates the required voltage to run the actuator. This voltage may be in the form of a 'Saw-Tooth' wave, or a ' Sine' wave or a ' Square' wave or any other form of electrical wave that would cause the piezoelectric bending actuator to bend up and/or down, thus moving the light source relative to the optical waveguide. Somewhere along the path of the l ight source, an overlap occurs between the optical mode of the light source and the optical mode of the optical waveguide, thereby generating a pulse of light traveling in the optical waveguide. [000205] The duration of the pulse is equal to the overlap time of the two optical modes. The duration of the pulse may be controlled either by changing one of the optical modes or by adjusting the scanning speed. The optical mode of the light source can be controlled by, for example, moving the lens in front of the emitter (see FIG. 2A) away or closer, thus expanding or converging the emitted light beam (i.e. the optical mode).
[000206] For example, in a system comprising a light source with a Gaussian shaped optical mode of 10 microns in width, and an optical waveguide with a Gaussian shaped optical mode of 1 micron in width, the width of the overlap of the two optical modes will be a convolution of the two, resulting in a Gaussian shaped optical mode of ~10-microns in width. If the piezoelectric bending actuator is driven with a periodic ' Saw-Tooth' wave having a frequency of 100 Hz with a scanning amplitude of 300 microns, the result is a 167 micro second pulse generated in the optical waveguide once every 5 milliseconds.
[000207] As with any other mechanical system, the piezoelectric bending actuator has limitations on its scanning speed. The components of the scanning-coupling system of the invention are therefore selected or designed to be able to meet the required scanning speed for a speci fic application. The current invention may be particularly advantageous in applications where the duration of the light pulses involved is of the order of a micro ( 10~6) second or longer.
[000208] A further embodiment of the invention is shown in FIG. 3 B, where the scanning-coupl ing system 300 consists of the piezoelectric bending actuator 305 and its electrical wires 306, the light source 302 and multiple optical waveguides 301 . The scanning light source will now generate trains of pulses in some or all of the optical waveguides based upon the chosen scanning path.
[000209] A further embodiment of the invention is shown in FIG. 3C. The scanning-coupling system 300 comprises multiple light sources 302, multiple optical waveguides 301 , a piezoelectric bending actuator 305 and its electrical wires 306. By selecting the properties of the different light sources (for example, their wavelength or optical mode.), it is possible to generate in each of the optical waveguides a train of pulses at many different wavelengths and with varying duration.
[000210] In a further embodiment depicted in FIG. 4A, the light source 402 is mounted on a rotating disk 403 to create a scanning-coupl ing system 400. The system further comprises an optical waveguide 401 . The rotating disk is driven by an electronic driver (not shown) causing the light source optical mode to scan the periphery of the disk through a point where it overlaps the optical mode of an optical waveguide. During this overlap, a light pulse is generated in the optical waveguide, the duration of which is equal to the duration of the overlap. In this case, the pulse duration is controlled by the shape of the two optical modes and the rotating speed of the disk.
1000211 ] A further embodiment of the scanning-coupl ing system 400 is shown in FIG. 4B.
Multiple light sources 402 are mounted on a disk 403 facing outwards, with multiple optical
waveguides 401 placed around the disk facing inwards. The time between two adjacent pulses in the same opt ical waveguide is controlled by the spacing between the l ight sources, as well as the rotation speed of the disk. There is also a well defined time correlation between the trains of pulses in all the optical waveguides.
[000212] In yet a further embodiment schematically shown in FIG. 5, the scanning-coupl ing system 500 consists of a piezoelectric bending actuator 505 and its electrical wires 506, mounted with a scanning lens 507. In this embodiment of the invention, the l ight source 502 and the optical
waveguide 501 are fixed. The scanning of the light beam emitted by the light source is achieved by moving the lens in front of the light source. The lens is disposed between the light source and the optical waveguide effective that movement of the actuator causes a light beam emitted by the light source to be directed by the lens effective to cause an overlap between the optical modes of the light beam and the optical waveguide.
[000213] FIG. 6 shows yet another non-limiting example of the scanning-coupling
system 600 where a stirring mirror 608 serves to deflect the light beam from light source 602 to scan space until it overlaps the Optical mode' of optical waveguide 601 . The light path 609 at one specific instant is also shown. The movement of the stirring mirror may be effected by any of the means disclosed herein, including, for example, a rotating disk, a motor, a solenoid, a hydraulic mechanism, a piezoelectric mechanism, a "memory-metal" mechanism, or a manually driven actuator.
[000214] In all of the above embodiments, the light source can be controlled by turning it 'on' and Off at chosen time periods, thus controlling which of the potential pulses is actually generated while the light sources are scanning through the optical waveguides. Furthermore, it should be understood that the scanning of the light source or its emitted light beam can be periodic, by constantly running the scanner in a periodic fashion, or it can be operated 'on demand,' generating anything from a single pulse to a large number of pulses.
[000215] The invention further comprises optical detection systems which utilize any of the scanning-coupling devices and methods described above. Additional components of such optical detection systems may include, for example, substrates for sample binding and/or processing, thermal transfer elements, thermistors, microchannels, reservoirs, electronic control boards, sample handl ing systems, an interface panel, and an enclosure or housing. FIG. 8 is a block diagram of a detection system representing one possible system where the scanning-coupling systems of the current invention may be used. Particular optical scanning systems where the scanning-coupling systems of the current invention are especial ly useful are described in U.S. Patent Publ ication Nos. 2007021 1 85, published Sep. 13, 2007, and 20090068668, published Mar. 1 2, 2009.
[000216] In various embodiments, the detection systems of the invention include a scanning l ight source and a substrate comprising a plurality of waveguides. The scanning l ight source emits one or more light beams that are spatially translated relative to the waveguides of the substrate such that the light beam is coupled to and in optical communication with the waveguides of the substrate at some point along its scanning path. By "spatially translated relative to the waveguides of the substrate" is meant that either the scanni ng l ight source is physical ly translated through space, or that the substrate comprising the waveguides is physical ly translated through space. (000217] In various embodiments, the scanning light source is a chip referred to herein as a "scanning light source chip." In embodiments where the scanning light source further incl udes detector elements, it may be referred to herein as a "scanning light source/detector,'' or a "scanning light source/detector chip." The scanning light source chips and scanning light source/detector chips of the invention are further referred to herein as "active scanning'1 chips in embodiments where the l ight generating elements (and detector elements when present) are integral to the chip, and as "passive scanning" chips in embodiments where the light generating elements (and detector elements when present) are external to the chip. The general term "scanning light source" may be used herein to encompass any or all of these embodiments of scanning light source chips and scanning light source/detector chips. Exemplary embodiments of systems of the invention including each of these types of scanning light source are described below.
[000218] FIG. 7A il lustrates an exemplary detection system 700 of the invention including an active scanning light source chip 702, a substrate 704, optical sensing sites 712 and a detector 706. The substrate includes excitation waveguides 708 and collection waveguides 710 that cross or intersect at an intersection region 714.
[000219] In one embodiment, as shown in FIG. 7A, the active scanning light source chip 702 is at some point along its scanning path coupled to and in optical communication with one or more of the excitation waveguides 708 at a first edge of the substrate704. Additionally, the detector 706 is coupled to and in optica! communication with one or more of the collection waveguides710 at a second edge of the substrate 704. Although a single detector at one edge of the substrate is shown, it is envisioned that two or more detectors could be coupled to and in optical communication with one or more collection waveguides or excitation waveguides at various edges of the substrate (not shown). For example, in one embodiment, where the active scanning light source chip is coupled to a first edge of the substrate, a first detector could be coupled to an adjacent edge and be in optical communication with a first end of a collection waveguide, whi le a second detector could be coupled to another adjacent edge and be in optical communication with a second end of a collection waveguide. A third detector can be coupled to the edge opposite to the one coupled to the active scanning light source chip and be in optical communication with the second end of the excitation waveguides (not shown).
[000220] As shown in FIG. 7A, in one embodiment the system 700 can be substantially planar. For example, the active scanning light source chip 702 can be a planar chip. This can be coupled to a planar substrate 704 that is a second chip that is further coupled to a planar detector 706 that is a third chip. In a particular embodiment, as shown in FIG. 7A, the system 700 is a planar lightwave circuit including three coupled chips. In one embodiment two chips are integrated into a single chip (e.g., the substrate chip and the detector chip). Such a configuration would be useful in a case where the substrate chip is reusable and can be effectively used for long periods of time. One appl ication of such a configuration would be in a system for detecting biological warfare-associated agents. In such an application it would be
advantageous for the system to operate for long periods of time without a need for replacing the chip. I n addition, having two chips integrated on a single substrate solves the problem of maintaining the relative alignment of two chips
[000221 ] Where the system is used in biological applications, including but not l imited to detection of biologically active analytes including nucleic acids, proteins or microorganisms, the substrate can be a multi-element bio-analysis chip.
[000222] In the embodiments of FIGS. 7A and 7C, it is envisioned that the crossing or intersecting of the excitation waveguides and the collection waveguides can be a direct physical crossing or intersecting, for example, where the excitation waveguides and the collection waveguides are embedded within the substrate in a single or in multiple layers. Alternatively, it is envisioned that the crossing or intersecting involves a physical space or distance between the excitation waveguides and the collection waveguides, for example, where the excitation waveguides and the collection waveguides are embedded within the substrate in separate layers. The optical sensing sites 712 of the system 700 typically are associated with the intersection regions714.
[000223] Typically one optical sensing site 712 is associated with each intersection region 714. As illustrated, in one embodiment the number of intersection regions 714 and optical sensing sites 712 is an arrangement of 100 intersection regions 714 and 100 optical sensing sites 712. It is envisioned that the number of intersection regions and optical sensing regions on a substrate chip can be greater than 10, greater than 100, greater than 1 ,000 or greater than 10,000. It is further envisioned that the density of intersection regions can be greater than 10 per cm2, greater than 100 per cm2, greater than 1 ,000 per cm2 or greater than 10,000 per cm2. In one embodiment the density of intersection regions is greater than 2,000 per cm2.
[000224] As further shown in FIGS. 7A and 7C, the crossing or intersecting of the excitation waveguides 708 and the collection waveguides 710 can be substantially perpendicular, for example, at an angle of 90°. Alternatively, in certain embodiments the crossing or intersecting can be angled less than or great than 90°.
[000225] It is also envisioned that in the embodiments of FIGS. 7A and 7C, a first light wave generated by the active scanning light source chip in an excitation waveguide induces the sensor to transduce an optical signal resulting in a second light wave in a collection waveguide, the second l ight wave being detectable by the detector.
[000226] As i llustrated in FIG. 7A, in one advantageous embodiment, the system 700 is a planar two-dimensional detection system. The system 700 in this embodiment includes a planar active scanning l ight source chip 702 comprising a plural ity of light source elements 71 8, for example an array of switchable lasers, which is scanned perpendicular to the plane of the substrate to couple at some point along its scanning path one or more pulses of light to the plane of the substrate 704. for example, a bio- analysis chip plane. Furthermore, the active scanning l ight source chip 702 can provide a dynamic source of light for selective and programmed excitation in respect to individual excitation waveguides 708. providing excitation to al l of the optical sensing sites 712 along that excitation waveguide 708. A dynamic light source includes but is not limited to a tunable wavelength and/or tunable bandwidth l ight source. Additional ly, the system 700 of this embodiment provides for planar col lection of the emitted light from al l the excited sensing sites 712 in the collection waveguides 710, specifically in the plane of the substrate 704, such that the l ight collection is substantially perpendicular to the direction of the light produced in the excitation waveguides 708.
[000227] In the embodiments of FIGS. 7B and 7D, it is envisioned that an optical sensing site 712 can be associated with each waveguide 708. It is envisioned that the number of optical sensing sites on a substrate chip can be greater than 10, greater than 100, greater than 200, greater than 1 ,000, greater than 5,000 or greater than 10,000. It is further envisioned that the density of optical sensing sites can be greater than 10 per cm2, greater than 100 per cm2, greater than 1 ,000 per cm2 or greater than 10,000 per cm2. In one embodiment the density of optical sensing sites is greater than 2,000 per cm2.
[000228] In the embodiments of FIGS. 7B and 7D, it is envisioned that a first light pulse generated by the scanning light source/detector chip in an in-coupling waveguide induces the sensor to transduce an optical signal resulting in a second light pulse in an out-coupling waveguide, the second light pulse being detectable by the detector.
[000229] FIG. 7B illustrates an exemplary detection system 700 of the invention including an active scanning light source/detector chip702,a substrate 704 and optical sensing sites 712. It is envisioned that the light source elements (718) can be any of a number of types of light sources including but not limited to switchable light sources or passive light sources. The active scanning light source/detector chip can include in-coupling waveguides 728, out-coupling waveguides 726 and combiners 730which combine the in-coupling and the out-coupling waveguides. Such combiners 730 are well known in the art. Substrate704 can include waveguides 708 and sensing sites 712 in relation to waveguides 708. For example sensing sites 712 can be on top of and in optical communication with waveguides 708. The active scanning light source/detector chip can include one or more detector elements 706.
|000230] In a second embodiment, as shown in FIG. 7B, light source elements 71 8 are coupled to and in optical communication with in-coupling waveguides 728 on the active scanning light
source/detector chip. Light generated by the light source travels along in-coupling waveguide 728 and is combined by combiner 730 into the out-coupling waveguide 726. The active scanning l ight
source/detector chip spatially scans perpendicular to the plane of the substrate through a point where each of waveguides 726 is coupled to and in optical communication with its counterpart waveguide 708 on the substrate 704. At this point a l ight pulse is generated in waveguide 708 travelling from left to right. The l ight pulse interacts with the sensing area712 which acts as a transducer to create a second light pulse travell ing in waveguide 708 from right to left. This second pulse couples to the out-coupling
waveguides 726 on the active scanning light source/detector chip. The second l ight pulse travels in waveguide 726 to the detector elements on the active scanning l ight source/detector chip.
[000231 ] FIG . 7C i llustrates an exemplary detection system 700 of the invent ion including a passive scanning l ight source 702, connected through optical fibers 720 to an external light source comprising light source elements 7 1 8, a substrate 704. optical sensi ng sites 71 2 and a detector 706. The passive light source chip 702 can include in-coupl ing waveguides or simply hold the end of the fibers. Substrate 704 includes excitation waveguides 708, collection waveguides 710 and sensing sites712 on top of and in optical communication with excitation waveguides 708 and collection waveguides 71 0.
Detector 706 can include one or more elements 716 as described herein.
[000232] In a third embodiment, as shown in FIG. 7C, light source elements 71 8 are coupled to and is in optical communication with the in-coupling waveguides of the passive scanning l ight source chip 702 through a set of optical fibers 720. Passive scanning light source chip 702 is at some point along its scanning path further coupled to and in optical communication with each of the excitation waveguides 708 at an edge of substrate 704. Additionally, detector 706 is coupled to and in optical communication with collection waveguides 710 at a second edge of substrate 704.
[000233] FIG. 7D illustrates an exemplary detection system 700 of the invention incl uding a passive scanning light source/detector chip 702, two sets of optical fibers 720, a light source comprising light source elements 718, a detector 706, a substrate 704, in-coupling waveguides 708 and 728, out- coupling waveguides 710 and 726 and optical sensing sites 712.
[000234] In a fourth embodiment, as shown in FIG. 7D, light source elements 71 8 are connected to and in optical communication with the in-coupling waveguides 728 on the passive scanning light source/detector chip 702 through a set of optical fibers 720. In addition, detector 106 is connected to and in optical communication with the out-coupling waveguides 726 on the passive scanning light source/detector chip through a second set of optical fibers 720. The passive scanning light source/detector chip, at some point along its scanning path, is coupled to and in optical communication with in-coupl ing waveguides 708 on the substrate 704. At that same point along the scanning path, the out-coupling waveguides 71 0 on substrate 704 are in optical communication with the out-coupl ing waveguides 726 on the passive scanning light source/detector chip.
[000235] FIG. 7E illustrates a side view of an exemplary detection system 700 of the invention including a scanning light source/detector chip 702, a substrate 704 and a piezoelectric bending actuator 705.
[000236] In the side view shown in FIG. 7E, the piezoelectric bending actuator is used to move the scanning light source/detector chip up and down thus scanning through a point along the scanning path where the scanning light source/detector chip is coupled to and in optical communication with the waveguides on substrate 704. At that point a first light pulse is generated in the in-coupling waveguides of substrate 704. At the same poi nt a second light pulse is coupled back from the out-coupl ing waveguides of substrate 704 to the out-coupling waveguides on the scanning l ight source/detector chip. While FIG . 7E illustrates the particular embodiment of the detection system shown in FIG . 7D, this particular embodiment is representative of any of the other embodiments of the detection system of the invention. Simi larly, the piezoelectric bending actuator is a representative of a variety of possible means for spatial ly translating the l ight emitted from the l ight source relative to the substrate. In various embodiments, the means for generating the relative motion of the light emitted from the l ight source relative to the substrate may incl ude a piezoelectric based motor, a step motor, an electrical motor, a magnetic actuator, a memory metal actuator, a solenoid, or a hydraulic actuator. The movement of the actuator may be effected by electrical power, thermal power, magnetic power or even mechanical power (i.e., manually).
[000237] FIGS. 7F and 7G illustrate another exemplary chip configuration where the one or more excitation waveguides 708 exit or extend to the same edge of the chip substrate as the one or more collection waveguides 71 0. As illustrated in FIG. 7F, the substrate has one excitation waveguide 708. In other embodiments, as illustrated in FIG. 7G, the substrate can have a plurality of excitation waveguides. The excitation waveguides 708 can cross the collection waveguides 710 as described herein. For example, the excitation waveguides 708 can cross the collection waveguides 710 perpendicularly or approximately perpendicularly to form a plurality of optical sensing sites 7 12 at the intersections, where each optical sensing site 712 is in optical communication with both the excitation waveguide 708 and the collection waveguide 71 0. The excitation waveguides 708 can bend or curve approximately 90 degrees to exit the same edge of the substrate as the collection waveguides 710. In some embodiments, the excitation waveguides 708 can be on one portion or side of the substrate while the collection waveguides 710 can be on another portion or side of the substrate. By having the excitation waveguides 708 exit the substrate at the same side as the collection waveguides 710, only one side or edge of the substrate or chip needs to be aligned with another chip or substrate having the light sources and detectors. By reducing the number of alignments needed between the components, the manufacturing process may be simplified, thereby reducing manufacturing costs.
[000238] FIG. 7H illustrates another embodiment of a chip configuration that is similar to the configuration shown in FIG. 7G, but with an added loopback waveguide 713 that also exits or extends to the same edge of the chip substrate as both the excitation waveguides 708 and collection waveguides 710. The loopback waveguide 71 3 can be an extension of one of the excitation waveguides 708 that loops back and extends to the edge of the chip substrate in which the other waveguides extend or exit. The loopback waveguide 713 can be used to efficiently align the chip with the l ight source, detectors, optical fibers, and/or other waveguides by detecting the injected l ight with a detector at the loopback waveguide output. The position of the chip can be adjusted until light is detected by the loopback waveguide detector. Without the loopback waveguide, very little light may enter the collection waveguides during alignment when no sample is present, making al ignment using the detectors more difficult.
[000239] In addition, FIG. 7H also illustrates an optical sensing zone 714 where the cladding has been removed to allow the light to interact with the optical sensing sites 71 2. In some embodiments as described elsewhere in this application, the cladding is only removed at the intersections between the collection and excitation waveguides where the optical sensing sites are located. However, in other embodiments, as shown in FIG. 7F1, a larger segment of cladding can be removed from the excitation waveguides, starting from a point before the excitation waveguides intersect the first col lection waveguides, extending across the col lection waveguides, and ending at a point after the excitation waveguides pass the last col lection waveguides. Light scattering can occur at the transition between a cladded and non-cladded portion of the excitation waveguide. This scattered l ight can enter the col lect ion waveguide and interfere with detection and measurements of light emission from the collection waveguide. By moving and eliminating the cladding transitions near the optical sensing sight, light scattering can be reduced, thereby improving the sensitivity and/or accuracy of the system. As shown, the cladding transitions for the optical sensing zones 714 are located away from the optical sensing sites and are limited to a single transition before the collection waveguides and a single transition after the collection waveguides. The cladding transitions of the optical sensing zones 714 can be located far enough away from the collection waveguides to prevent, minimize or reduce light scattering interference.
[000240] Although four exemplary embodiments of the detection system of the invention are specifically disclosed herein, it is envisioned that any of a number of other combinations of coupling the different components/chips disclosed herein at different edges of the components/chips are possible. For example, in one embodiment, a first scanning light source/detector chip is coupled to a first edge of the substrate and a second scanning light source/detector chip is coupled at a second edge of the substrate (not shown). It can be understood accordingly that the passage of light pulses within the devices and systems described herein, though described in terms of "left" and "right" can be practiced in a variety of directions and orientations based on the flexible arrangements of components provided herein.
Furthermore, additional combinations of the different scanning chips with the different substrates are envisioned. For example, the scanning light source chips shown in FIGS. 7A and 7C may be used in combination with the substrates shown in FIGS. 7B and 7D, together with a detector in optical communication with an opposite end of the substrate. In addition, in the embodiments illustrated in FIGS. 7B and 7D, either the scanning chip or the substrate may comprise at least one combiner.
[000241] Although in all of the above embodiments the scanning light source is spatially translated relative to the substrate, it is further envisioned that the scanning of the emitted l ight relative to the waveguides of the substrate may also be effected by spatially translating the substrate relative to the light source, or by spatially translating any part or component of the scanning l ight source, such as one or more mirrors, lenses, or prisms, using any of the means disclosed herei n.
[000242] It is envisioned that an optical sensing site 712 can be associated with each
waveguide 708. It is envisioned that the number of optical sensing sites on a substrate chip can be greater than 10, greater than 100, greater than 200, greater than 1 ,000, great than 5,000 or greater than 10,000. It is further envisioned that the density of optical sensing sites can be greater than 10 per cm", greater than 1 00 per cm2, greater than 1 ,000 per cm2 or greater than 10,000 per cm2. In one embodi ment the density of optical sensing sites is greater than 2,000 per cm2.
[000243] It is envisioned that in any of the embodiments described herein, that a first light pulse generated by the scanning light source chip in an in-coupling or excitation waveguide induces the sensor to transduce an optical signal resulting in a second l ight wave in an out-coupling or col lection waveguide, the second light wave being detectable by the detector.
[000244] FIG. 8 is an exemplary illustration of the detection system of the invention as part of a working system 801 in a housing 809. Whi le the detection system i l l ustrated in FIG . 7A-7E is the core of the present invention, in order to facilitate the operation of this system, one or more other modules can be included in a working system that includes the detection system components of the invention.
[000245] FIG. 8 i l lustrates one possible configuration for a working system 801 that can include housing 809 for enclosing various modules of working system 801 including but not limited to substrate 804, robotic system 803, scanning light source chip802, multi-element detector 806, electronic boards 807 and interface panel 805. Substrate 804, scanning l ight source chip802, and multi-element detector 806, are discussed in detail below.
[000246] In regard to housing 809, as shown in FIG. 8, in one embodiment an enclosure or housing 809 holds in place two fixed chips (e.g., of a 3-chip architecture), namely, scanning light source chip 802 and multi-element detector 806. Accordingly, in this embodiment substrate chip 804 is movable in relation to scanning light source chip 802 and multi-element detector 806. Housing 809 can include any number of accurately machined parts and or components as described herein, allowing, for example, the relative alignment of the 3 optical chips, and the movement of the scanning light source chip perpendicular to the plane of the substrate. The working system housing can optionally include temperature control and vibration isolation for the working system (not shown).
[000247] As shown in FIG. 8, working system 801 can further include an X, Y, Z, Θ robotic system 803 for positioning substrate 804 as required within working system 801 . X, Y, Z, Θ robotic system 803 can be a translation stage with several degrees of freedom for receiving or accepting substrate 804, holding it in place, and aligning it in relation to the rest of working system 801 . As desired, at the end of a run X, Y, Z, Θ robotic system 803 can eject substrate 804 from working system 801 .
[000248] It is envisioned that the working system can further include an al igning system (not shown). An aligning system can include one or more light sources, one or more detectors and one or more cameras for active detection of the position of the substrate of the invention. Based on the detected position, the aligning system can align the substrate to the rest of the working system modules, for example, to provide aligned optical communication between the substrate and the scanning light source/detector chip.
[000249] As shown in FIG. 8 working system 801 can further incl ude one or more electronic boards 807, for example, an electronic driving board and a control board. It is envisioned that one or more electronic boards can control all the different parts of the working system. Electronic boards 807 can control scanning light source 802 and any other light source present in the system. Electronic boards 807 can be adapted to read any or all of the detectors and cameras in working system 801 .
Electronic boards 807 can further be adapted to drive robotic system 803 and control its motion, to control the motion of the scanning light source chip, and optionally to monitor and control temperature in different areas of the system. Electronic boards can incl ude logic elements and processors (not shown). It is envisioned that electronic boards can further include embedded software both for control ling the working system and for interfacing the outside world, for example by way of interface panel 805 which can include a key-pad or any other input/output port. [000250] As shown in FIG. 8 working system 801 can additional ly incl ude one or more interface panel 805. It is anticipated that the system wi ll have one or more interface panel 805 to allow a user to interface with the system and operate it. Interface panels can include any number of input and output ports well known in the art for connecting the system to other systems or to an external control console (not shown).
|000251 ] FIG. 9A illustrates an exemplary substrate 904 of the second embodiment of the detection system of the current invention (as show in FIG. 7B) further including barriers 91 1 intended to block stray light within the substrate and reduce crosstalk between the different elements of the substrate. Barriers 91 1 can be light absorbing or light reflecting. Barriers 91 1 can be variously sized, shaped and positioned between waveguides 908 in any of a number of orientations to achieve a desired optical effect. As shown in FIG. 9A, barriers 91 1 can be arranged between two adjacent waveguides and proximal to optical sensing sites912. Waveguides 908 are used to guide a primary l ight wave (excitation light; see dashed arrow) from the left edge of substrate 904 to optical sensing sites 912. Waveguides 908 then guide a secondary light wave (collected at optical sensing sites 912; see dashed arrow) from optical sensing sites 912 back to the left edge of substrate 904.
[000252] FIG. 9B illustrates an exemplary substrate 904 of the fourth embodiment of the detection system of the current invention (as shown in FIG. 7D) further including in-coupl ing waveguides 908 and combiners 926. The primary light wave (excitation light; see dashed arrow) is coupled to
substrate 904 through in-coupling waveguides 908 at the left edge of substrate 904. The excitation light traveling from left to right is combined by combiners 926 into out-coupl ing waveguides 910 which further guide it to optical sensing sites 912. Out-coupling waveguides 910 are then used to guide the secondary light wave (collected at sensing sites 912; see dashed arrow) from optical sensing
sites 912 back to the left edge of substrate 904. Barriers 1 1 have the same purpose as described above in FIG. 9A.
1000253] FIG. 9C schematical ly il l ustrates a cross section of the substrate 904 of one embodiment of the current invention. In the example illustrated, in/out coupling waveguides 908 are embedded underneath a surface of substrate 904. Optical sensing sites 912 can be etched into a surface, for example, the upper cladding of substrate 904 and located, for example, ad jacent to and on top of
waveguide 908 facil itating optical communication between them. It is envisioned that in a different embodiment, the optical sensing sites can also be located on the surface of substrate 904 or etched only part of the way into the upper cladding, or all the way through the waveguides (not shown). It is also envisioned that the waveguides can be single-mode waveguides, multi-mode waveguides or any combination of the two, namely, single mode in the vertical dimension and multi-mode in the lateral dimension.
[000254] FIG. 9D i llustrates an exemplary substrate 904 of the first and third embodiments of the detection systems of the current invention (as shown in FIGS. 7A and 7C), further including
barriers 91 1 intended to block stray l ight withi n the substrate and reduce crosstal k between the different elements of the substrate. The barriers 91 1 can be l ight absorbi ng or l ight reflecting. The barriers 91 1 can be variously sized, shaped and positioned between the col lection waveguides 91 0 and/or the excitation waveguides 908 in any of a number of orientations to achieve a desired optical effect. As shown in FIG. 9D, the barriers 91 l ean be arranged in a row between two adjacent col lection waveguides and proximal to the optical sensing sites 912 and intersection region 914.
[000255] As shown in FIG. 9E, (in this view a top cladding layer is not shown) in one embodiment the substrate 904 can include excitation waveguides 908 and col lection waveguides 910 embedded beneath a surface of the substrate 904 in multiple layers. As shown, the excitation waveguides 908 cross, physically intersect, and are in optical communication with the collection waveguides 910 at the intersection regions 914. In the embodiment shown in FIG . 9E, the optical sensing site 912is positioned at the intersection region 914 above and in optical communication with the excitation waveguides 908. As further shown in FIG. 9E, the substrate 904 includes multiple layers including a silicon layer 920 and a silica (Si02) layer 922, wherein the excitation waveguides 908 and the collection waveguides 910 are embedded within the silica (Si02) layer 922.
[000256] As shown in FIG. 9F, in another embodiment the substrate can include excitation waveguides 908 and collection waveguides910 embedded underneath a surface of the substrate 904 in a single layer. As shown, the excitation waveguides 908 cross, physically intersect and are in optical communication with the collection waveguides 910. In contrast to the embodiment shown in FIG. 9E, here the intersection between excitation waveguides 908 and collection waveguides 910 occurs internal to the collection waveguides 91 0. As further shown in FIG. 9F, the substrate 904 includes multiple layers including a sil icon layer 920, a silica (Si02) layer 922, and a cladding layer 924. As shown, the excitation waveguides 908 and collection waveguides 910 can be embedded within the silica (Si02) layer 922. Additionally, the optical sensing site 912 can be embedded within both the cladding layer 924 and the silica (Si02) layer 922. Optional ly, the optical sensing site can be embedded solely within the cladding layer (not shown).
[000257] It is envisioned that the excitation waveguides and collection wave guides can be single- mode or multi-mode waveguides. In one embodiment, the excitation waveguides are single-mode and the collection waveguides are multi-mode. It is envisioned that waveguide configurations can include single- or multi-mode configurations in either vertical or lateral orientations within a waveguide. For example, in one specific and non-l imiting embodiment, the excitation waveguides 908 can support a single mode in the vertical dimension and multi modes in the lateral dimension. Optional ly, as shown in FIG. 9D, the excitation waveguides 908 and the col lection waveguides 910 can span the entire substrate from one edge to another edge.
[000258] As shown in FIG. 9F. the substrate 904 components and optical sensing sites 912 can include dimensions. FIG. 9F shows two cross-section views of the substrate 904. View AA is a cross- section view in plane A as indicated in FIG. 9D and FIG. 9E. View BB is a cross-section view in plane B as i ndicated in FIG. 9D and FIG . 9E. As shown in FIG. 9F. the thickness of the cladding layer 924 above the excitation waveguides can be about 0. 1 μιπ to about 20 μιη. In one embodiment the cladding layer 924 thickness is about 1 μηι to about 2 μιη. By way of a non-l i miting example, as shown in FIG. 9F, an opening of the optical sensing site 912 can include the following dimensions: about 20 μιη by about 2 μηι. The distance between collection waveguides 910 can range from about 1 μιη to about 1000 μιη. For example, as shown i n FIG. 9F, the distance between collection waveguides 910 can be about 100 μιτι. The distance between collection waveguides 910 and the silicon layer 920can be about 1 μιη to about 1 00 μιη. For example, as shown in FIG. 9F. the distance between collection waveguides 910 and the silicon layer 920 can be about 10 μηι to about 20 μιη.
[000259] As shown in FIGS. 9E and 9F, the excitation waveguides 908 and collection waveguides 910 can be channel waveguides. Exemplary ranges for waveguide dimensions in the embodiment shown in FIGS. 9E and 9F include about 0. 1 to about 1 0 μιη thick and about 1 to about 100 μιτι wide. By way of non-limiting example only, the excitation waveguides 908 can include cross-section dimensions of about 0.1 μιη by about 100 μιη and the collection waveguides 910 can include cross- section dimensions of about 0.2 μιτι by about 1 00 μιπ.
[000260] FIG. 9G in a side view illustrates another embodiment of substrate 904 of the invention in relation to a thermal transfer element 903, for example, a thermoelectric cooler (TEC). Thermal transfer element 903 is a temperature control system useful for heating or cooling a chip, for example, substrate 904. Although the thermal transfer element may be referred to herein as a cooling element, it is to be understood that where the thermal transfer element is configured to increase and decrease the temperature of a chip, the component functions essentially as a heating and as a cooling element depending on the induced direction of the electrical current. The thermal transfer element can provide a range of useful temperatures. For example, the thermal transfer element can be configured to provide a temperature in the range between about -40° C. to about 120° C. as desired. The thermal
transfer 903 element can be adapted to receive substrate 904 of the invention. The thermal transfer element 903 can be adapted to contact part or al l of a surface of the substrate 904 of the invention.
[000261] Providing thermal transfer element 903 in conj unction with substrate 904 of the invention is useful, for example, for the amplification of tested sample molecules through processes such as the polymerase chain reaction (PCR) as described herein. In use, the embodiment as described for FIG. 9G provides the capability of controll ing the temperature of the entire substrate such that as the temperature of the entire substrate is cycled, samples at any optical sensing site can be amplified by PCR simultaneously.
[000262] FIG. 9H illustrates another embodiment of substrate 904 of the invention wherein optical sensi ng site 91 2 includes heater 905and thermistor 907. In this embodiment, optical sensing site 912 of substrate 904 can include heater 905, for example, a thin-film heater, in the vicinity of each sensing sites 912. Heater 905 can be adapted to enable individual temperature control for each sensing site 912. In addition to heater 905, thermistor 907 can be located at or near each sensing site 91 2 thereby providing for measuring the local temperature. In use. this embodiment provides the capability of running the same or any desired different number of cycles and the same or any desired different temperature profiles for each and every sensing site. [000263] FIG. 91 i l lustrates yet another embodiment of substrate 904 of the invention wherein optical sensing site 912 includes heater905 and thermistor 907. In this embodiment, optical sensing site 912 of substrate 904 can include heater 905, for example, a thin-fi lm heater, in the vicinity of one or more sensing site 912. Heater 905 can be adapted to enable individual temperature control for each sensing site 912. In addition to heater 905, thermistor 907 can be located at or near one or more sensing site9 ! 2 thereby providing for measuring the local temperature. In use, this embodiment provides the capability of running the same or any desired different number of cycles and the same or any desired different temperature profiles for each and every sensing site.
[000264] Advantageously, the embodiments described for FIGS. 9G, 9H, and 91 can support real- time PCR. As described herein, since optical detection is done from within the substrate, signal detection these both embodiments (see FIGS. 9G, 9H, and 91) can be done while the samples are in the process of the amplification cycles, thereby enabling real time analysis of the PCR process.
[000265] FIG. 9J illustrates yet another embodiment of substrate 904 of the invention wherein substrate 904 additionally includes reservoirs 913 and microchannels 909 in relation to optical sensing sites 912. As such, in this embodiment microfluidics are incorporated into the substrate. Microfluidics can be adapted to drive liquid (in this case the tested sample) using the capillary effect across the substrate. As illustrated in FIG. 9J, this can be achieved by an arrangement of microchannels 909, optionally of varying width, which force the sample from one or more reservoirs 913 to optical sensing sites 912 which can include etched wells to receive the sample. The microchannels can be either etched on the face of the chip itself or can be added as an external structure on a surface of the substrate.
[000266] FIG. 9K illustrates yet another embodiment of substrate 904 of the invention wherein substrate 904 additionally includes reservoirs 91 3 and microchannels 909 in relation to optical sensing sites 912. As such, in this embodiment microfl uidics are incorporated into the substrate. Microfluidics can be adapted to drive liquid (in this case the tested sample) using the capi llary effect across the substrate. As i llustrated in FIG. 9K, this can be achieved by an arrangement of microchannels 909, optionally of varying width, which force the sample from one or more reservoirs 913 to optical sensing sites 912 which can include etched wells to receive the sample. The microchannels can be either etched on the face of the chip itself or can be added as an external structure on a surface of the substrate 904.
[000267] In use, it is envisioned that a sample to be tested can be pipetted into a reservoir at one end of the substrate. The sample can then be distributed using the microfluidic system to the optical sensing sites and sensing wells where it is al lowed to bind to pre-spotted probes and can subsequently be optically detected and analyzed. Several reservoirs may be used to separate different samples/patients or for running several parallel tests.
[000268] The substrate of the system may be dip-coated with one or more probes configured to interact biochemical ly with a desired biological ly active analyte molecule. Example 1 describes chip coating protocols for antibody or ol igonucleotide attachment.
[000269] Additional ly, it is envisioned that one or more probes may be appl ied to a sensor of the optical sensi ng sites using a print head. Furthermore, it is envisioned that delivery of sample to the optical sensing sites of the system comprises del ivering the sample using an assay head. One possible print head technology is described in U.S. patent application Ser. No. 1 1 /241 ,060, filed on Sep. 30, 2005, and U.S. patent application Ser. No. 1 1 /632,086, filed on Jul . 6, 2005.
[000270] FIG. 10A in a top view illustrates an exemplary substrate 1004 of the system of the invention wherein the collection waveguides 1010 include funnels 1017 (shown in detai l in FIG. 10B) for collecting light.
[000271 ] As shown in the example in FIG. 10A, the substrate 1004 can include a 10x 10 array consisting of 10 excitation waveguides 1008 (e.g., about 5 μιη wide by about 2 μιη deep), 10 collection waveguides 1010 (e.g., about 30 μιη wide by about 1 0 μιη deep), 100 optical sensing sites 1012 (e.g., wel ls about 30 μιη long by about 5 μιη wide by about 10 μιη deep), 100 funnelsl 017 for collecting light from the optical sensing sites 1012 and barriers 101 1 (e.g., light absorbing channels) to reduce crosstalk between the optical sensing sites 1012. Although the example shown in FIG. 10A includes a 10* 10 array of excitation waveguides 1008 and collection waveguides 1010, it is envisioned that the substrate can include greater than 10, greater than 100 or greater than 1 ,000 excitation waveguides 1008 and collection waveguides 1010.
[000272] In the embodiment shown in FIG. 10A, excitation light can be coupled into one or more excitation waveguides 1008 on the left hand side of the substrate 1004 from, for example, a scanning light source chip. Excitation l ight can travel along the excitation waveguides 1008 and couple into the optical sensing sites (e.g., wells) through an evanescent field tail. Excited fluorescence generated in the optical sensing site 1012 can be col lected along the long facet of the optical sensing site 1012into the funnels 1017. The funnels 1 017 can channel the light into the collection waveguides 1010. The light in the collection waveguides 101 0 can be coupled out at the "bottom" of the substrate 1004 into a detector array (not shown). Light scattered outside the optical sensing sites 1012 can be blocked by a series of barriers 1 01 1 (e.g., light absorbers) to avoid crosstalk between parallel collection waveguides 1010.
[000273] In one embodiment, the substrate shown in FIG. 10A includes two waveguide layers. As illustrated in cross-sectional view inFIG. I OC, a bottom layer, about 2 μηι thick, can include the excitation waveguide 1008. The bottom layer can have a higher refractive index in order to increase the evanescent field tail presence in the optical sensing sites. An upper layer, about 10 μιτι thick, can contain the optical sensing site and the light collection structures (funnels and waveguides). The upper layer can have a lower refractive index than the bottom layer in order to minimize l ight loss when coupling the light out of the substrate to the detector.
[000274] In a particular embodiment of the above, both the excitation and collection waveguides are multimode.
[000275] As shown in cross-sectional view in FIG. I OC, in order to minimize the loss of l ight at the waveguide crossing points due to light coupl ing from the col lection waveguides 1010 into the excitation waveguides 1 008. the excitation waveguides 1 008 can be thinner than the collection waveguides 1 010. For example, as shown in F IGS. 1 013 and I OC, the excitation waveguides 1008 can have a width of about 5 μιη (see FIG. 1 0B) and a height of about 2 μηι (see FIG. I OC). As further shown, the col lection waveguides 1010 can have a width of about 30 μιη (see FIGS. 10A and 10B) and a height of about 1 0 μπι (see FIG. I OC).
[000276] It is envisioned that light coupled at the waveguide crossing points between the excitation waveguides and the collection waveguides can shine directly into the optical sensing sites, thereby increasing light excitation rather than being lost.
[000277] As shown in FIG. 1 0B, the optical sensing sites can be wells that are narrow (about 5 μηη) and long (about 30 μιη) with light collectable along the long facet. Such a configuration increases the efficiency of light collection. In addition, light excitation coupling into the well can increase due to the long coupling length. The wel l dimensions (5*30x 10 μηι3) yield a volume of 1 .5 pico-liter. Larger wells are also envisioned in a variety of sizes yielding volumes ranging from about 0.1 pico-liter to about 100 micro-liter.
[000278] The funnel can have a radius for the collection, confinement and coupling of light into the collection waveguides. Suitable ranges for radii can include from about 100 μιη to about 1000 μιη.
[000279] The barriers 101 1 as illustrated in FIGS. 10A and 10B, can be trenches filled with light absorbing material (e.g., a metal such as gold). Where the barriers 101 1 are trenches, the trenches can include openings above the excitation waveguide 1008 to avoid loss at the crossing points (not shown).
[000280] The overall dimensions of the substrate illustrated in FIG. 10A can be about 1.2 by about 1 .2 mm2. Margins can optionally be included around the substrate to adjust the overall dimensions as desired.
[000281] FIG. 1 1 A i llustrates an exemplary substrate 1 104 of the system of the invention wherein the excitation waveguides 1 ! 08include a plurality of branches 1 121 (shown in detail in FIG. 1 I B) for tapping light from the excitation waveguides and coupling it into the sensing wells.
[000282] In the embodiment shown in FIG. 1 1 A, the substrate 1 104 can be made up of several waveguide layers (e.g.. three waveguide layers). Such a configuration can be useful, for example, to optimize excitation and fl uorescence collection while minimizing loss and crosstalk. FIGS. 1 1 C and 1 1 D are schematic cross-section views of the substrate 1 104 through planes at (AA) and (BB) respectively as indicated in FIG. 1 1 B.
[000283] In one embodiment the substrate consists of three waveguide layers having core refractive index of 1 .7 and clad reflective index of 1 .4. Useful core refractive index values range from about 1 .45 to about 2. 1 . and useful clad refractive i ndex val ues range from about 1 .4 to about 1 .5.
[000284] As shown in FIGS. 1 1 C and 1 I D, in one embodiment where the substrate 1 104 includes three waveguide layers, a first bottom layer can be about 10 μιη thick and include the collection waveguides 1 1 10. In the embodiment il lustrated in FIG. 1 1 A, the collection waveguides 1 1 10 can be about 30 μιη wide, multimode and traverse the substrate 1 104 from substantially edge to edge. A second middle waveguide layer can be about 0.5 μηι to about 1 μιη thick and include coupl ing waveguide branches 1 1 21 (see FIGS. 1 0A and 10B). The branches 1 121 can couple excited light into the optical sensing sites, which can be wel ls. A third top layer can be about 2 μιη thick and include single-mode excitation waveguides 1 1 08 and traverse the substrate 1 104 substantially from edge to edge. [000285) FIG. 14A il lustrates an active scanning light source chip 1402 of the first embodiment (see FIG. 7A) of the detection system of the current invention. The active scanning light source chip 1402 i ncludes l ight source elements 141 8 and in-coupling waveguides 1440. A primary light wave generated by the light source elements is coupled from left into in-coupling waveguides 1440.
Waveguides 1440 guide the primary light wave to the right edge of the active scanning light source chipl 402 and couple it out to the substrate (not shown).
[000286] FIG. 14B i llustrates an active scanning light source/detector chip 1402 of the second embodiment (see FIG. 7B) of the detection system of the current invention. The active scanning light source/detector chip 1402 includes light source elements l 418, detector elements 1416, in-coupling waveguides 1440, out-coupling waveguides 1438 and combiner 1436. A primary light wave (excitation light) generated by the light source 1418 is coupled from left into in-coupling waveguides 1440. The excitation light is then combined by combiners 1436 into out-coupling waveguides 1438 which then guide it to the right edge of active scanning light source/detector chip 1402 and couple it out to the substrate (not shown). The substrate (not shown) couples back the secondary light wave (collected at the optical sensing sites— not shown) to out-coupling waveguides 1438at the right edge of active scanning light source/detector chip 1402. The secondary light wave is guided by the out-coupling
waveguides 1438 to the detector elements 1416 on the active scanning light source/detector chip.
[000287] FIG. 12A illustrates a passive scanning light source chip 1202 of the third embodiment (see FIG. 7C) of the detection system of the current invention. Passive scanning light source
chip 1202 includes in-coupling waveguides 1228. A primary light wave (excitation light) is coupled from left into in-coupling waveguides 1228 through a set of optical fibers (not shown). Waveguide l 228 guides the primary light wave to the right edge of the passive scanning light source chip 1202 and couples it out to the substrate (not shown).
[000288] FIG. 12B il lustrates a passive scanning light source/detector chip 1202 of the fourth embodiment (see FIG. 7D) of the detection system of the current invention. The passive scanning light source/detector chip 1202 includes in-coupling waveguides 1228, out-coupling waveguides 1226 and combiners 1230. A primary l ight wave (excitation light) is coupled from the left into in-coupling waveguides 1228 through a set of optical fibers (not shown). The excitation light is then combined by combiners 1230 into the out-coupling waveguides 1226 which then guide it to the right edge of passive scanning l ight source/detector chip 1 202 and couple it out to the substrate (not shown). The substrate (not shown) couples back the secondary light wave (col lected at the optical sensing sites— not shown) to out- coupling waveguides 1226 at the right edge of passive scanning light source/detector chip 1202. The secondary l ight wave is guided by waveguides 1 226 from right to left and is coupled out of passive scanning l ight source/detector chip 1202 at its left edge to the detector (not shown) through a set of optical fi bers (not shown).
|000289] FIG . 12C illustrates a second possible design of a passive scanning light source/detector chip 1202 of the fourth embod iment (see FIG. 7D) of the detection system of the current invention. The passive scanning l ight source/detector chip 1202 includes in-coupl ing waveguides 1 228 and out-coupling waveguides 1226. A primary l ight wave (excitation l ight) is coupled from left into in-coupling waveguides 1228 through a set of optical fibers (not shown). The excitation light is then guided to the right edge of passive scanning light source/detector chip 1202 where it is coupled out to the substrate (not shown). The substrate (not shown) couples back the secondary light wave (collected at the optical sensing sites— not shown) to out-coupl ing waveguides 1226 at the right edge of passive scanning light source/detector chip 1202. The secondary light wave is guided by waveguides 1226 from right to left and is coupled out of passive scanning l ight source/detector chip 1202 at its left edge to the detector (not shown) through a set of optical fibers (not shown).
[000290] It is envisioned that the light source elements 718 in FIGS. 7A- 1 D (or 1418 in FIGS. 14A and 1 4B) can comprise a dynamic light source allowing for selective and programmed generation of the primary light wave in one or more individual elements.
[000291] In some embodiments, the light source elements 718 can provide variable wavelengths of light. In one embodiment, the light source element is a broad-band source. In another embodiment, the light source element is a tunable source.
[000292] In some embodiments, the number of light source elements 718 will be equal to the number of excitation waveguides in the substrate of the system. The interface between the scanning light source outputs should match, in terms of pitch, the excitation waveguides in the substrate to allow these two elements at some point along the scanning path to efficiently couple and transfer light from the scanning light source chip to the excitation waveguides or to the in-coupling waveguides of the substrate.
[000293] The light source elements depicted in FIGS. 7A-D and FIGS. 14A and 14B can include different types of light generating elements. In some embodiments, the light generator elements are light emitti ng diodes (LEDs). In other embodiments the light generator elements are laser diodes (LDs). Each individual l ight generator element is separately controlled and can be turned on or off as desired. In one embodiment the scanning l ight source chip includes 10 or more light generator elements. In another embodiment the scanning l ight source chip includes 1 00 or more light generator elements. In yet another embodiment the scanning light source chip includes 1000 or more light generator elements. In a further embodiment the scanning light source chip includes between 10 and 100 light generator elements.
[000294] The detector elements depicted in FIGS. 7A-D and FIGS. 14A and 14B can include different types of detector elements. In some embodiments, the detector elements are PIN diodes. In some embodiments the detector elements are avalanche photo-diodes (APD). In some embodiments the detector elements are a group of pixels which are part of a CCD array. Each individual detector element is separately controlled and read. In one embodiment the scanning light source chip includes 10 or more detector elements. In another embodiment the scanning light source chip includes 100 or more detector elements. In yet another embodiment the scanning light source chip i ncludes 1000 or more detector elements. In a further embodiment the scanning l ight source chip includes between 1 0 and 100 detector elements. [000295] In one non-l imiting example, the detector element has a spectral range of between 400 to 1 000 nm, a photosensitivity (A/W) of >0.3, an active area per element of 0.005 mm2, 128 elements, and a pitch of <0.1 mm.
[000296] In one embodiment, the detector is a silicon photodiode (PN, PIN, CCD or APD) array. An example of a suitable detector array is the Hamamatsu 64x2048 CCD chip (PN— S I 0420-1 106).
[000297] In some embodiments, the light source elements and detector elements on the scanning l ight source chip can be integrated on a single chip which includes an array of two or more light source elements, an array of two or more detector elements, an array of two or more in-coupling waveguides, an array of two or more out-coupling waveguides and an array of two or more combiners. In one implementation each light source element is optically coupled to one in-coupling waveguide and adapted such that most of the l ight emitted by the light generator element propagates along that waveguide. The waveguides can extend to the edge of the chip where they can be brought at some point along the scanning path of the chip to couple the light propagating within them to the substrate. In one implementation two light source elements, each optionally emitting at a different wavelength can be coupled to a single in-coupling waveguide. In another implementation more than two light source elements, each optionally emitting at a different wavelength can be coupled to a single in-coupling waveguide.
[000298] In other embodiments, the light source elements on the scanning light source chip can be integrated on a single chip which includes an array of two or more light source elements and an array of two or more waveguides. In one implementation each light source elements is optically coupled to one waveguide and adapted such that most of the light emitted by the light source element propagates along the waveguide. The waveguides can extend to the edge of the chip where they can be brought to couple the light propagating within them to the substrate. In one implementation two light source elements, each optionally emitting at a different wavelength can be coupled to a single waveguide. In another implementation more than two l ight source elements, each optionally emitting at a different wavelength can be coupled to a single waveguide.
[000299] The scanning l ight source chip can include, in addition to light source elements, detector elements and waveguides, light manipulating features such as lenses, filters, switches, modulators, spl itters, combiners, mirrors and circulators.
|000300] The control of the scanning light source chip can be either integrated on the same chip as the l ight source elements, detector elements and waveguides or alternatively can be external to the chip. The scanning l ight source chip can have an electrical interface to an external driver or external controller or logic interface to an external control system. The control of the light source elements and detector elements al lows driving each l ight source element and each detector element separately. It further al lows also control of the other features present on the scanning l ight source chi p such as, for example, the modulators and switches.
1000301 ] Additional elements useful in planar l ightwave circuits, incl uding but not limited to couplers, fi lters, mirrors, circulators, splitters, modulators, switches and trenches are envisioned as part of the system described herein (not shown). Such elements when integrated into the substrate or into the scanning light source chip can serve to manipulate the incoming first light waves in the in-coupl ing waveguides or outgoing second light waves in the out-coupling waveguides. In other embodiments, such elements when integrated into the substrate or into the scanning l ight source chip can serve to manipulate both incoming first l ight waves in the excitation waveguides or outgoing second light waves in the collection waveguides.
[000302] A range of dimensions for the various features described herein include: waveguides thickness— about 20 mil to about 50 μιη; waveguide width— about 1 μιη to about 500 μχη; waveguide length— about 1 mm to about 100 mm; optical sensing site length— about 100 μιη to about 100 mm; optical sensing site width— about 1 μιιη to about 500 μηι; optical sensing site depth— about 0 μιη to about 20 μιτι; waveguide pitch— about 10 μηι to about 10 mm; substrate thickness— about 1 00 μιη to about 5 mm; upper cladding thickness— about 0 μιτι to about 20 μιπ; and lower cladding thickness— about 0.1 μιη to about 20 μιη.
[000303] The substrate of the detection system can made up of any of a number of well known materials suitable for use in planar lightwave circuits. For example, useful substrate materials include but are not limited to silica (Si02), glass, epoxy, lithium niobate and indium phosphide as wel l as combinations thereof. The waveguides disclosed herein can be made up of silicon, silica (Si02) and derivatives thereof, si licon oxynitride (SiON) and derivatives thereof, silicon nitride (SiN) and derivatives thereof, tantalum oxide (TaOx) and its derivatives thereof, polymers, lithium niobate and indium phosphide as well as combinations thereof. In one embodiment, UV light is used to change the refractive index of a waveguide material after deposition.
[000304] FIG. 13A i llustrates an exemplary sil icon layer 1326 of the substrate 1304. For example, the silicon layer 1326 can be made up of a silicon wafer having a thickness from about 0.1 mm to about 1 0 mm. In another example the silicon wafer can have a thickness from about 0.3 to about 1 mm. In a particular example as ill ustrated in FIG. 13A, the silicon wafer has a thickness of 0.65 mm. As shown in FIG. 13A in one embodiment, the silica (Si02) layer 1322 is a 14 μιη thermal oxide layer of silica (Si02) created by placing the silicon in an oxygen-rich environment inside a furnace at high temperature. The top sil icon layer oxidizes over time (several hours) creating a Si02 layer. Additionally, as shown in FIG. 13 A, in one embodiment, the cladding layer 1324 is 15 μιη thick and deposited by a PECVD (Plasma-Enhanced Chemical Vapor Deposition) process after etching to produce the waveguides 1308.
[000305] It is envisioned that the various layers of the substrate can include different refraction index properties. For example, a waveguide layer (e.g. SiN) has a higher refraction index than a cladding layer of si l ica deposited thereon.
[000306] As shown in FIG. 13 B (i llustrated with a photomicrograph prior to deposition of a cladding layer), in some embodiments, the substrate 1304 can include two waveguides 1 308 arranged for l ight wave coupling on a si lica (Si02) layer 1322. Alternatively, as shown in FIG. 13C. two
waveguides 1 308 can be arranged for guiding uncoupled light waves on a si lica (Si02) layer 1 322and over-clad with a cladding layer 1 324. [000307] The optical sensing sites in one embodiment are in the form of wells, for example, etched wel ls (see FIG. 9C cross-section view). Where the optical sensing site is a well, it can act as a vessel for a l iquid sample. In another embodiment the optical sensing sites are a region on the surface of the substrate, for example, above the waveguides. In a further embodiment, the optical sensing sites are biochemical interaction sites. For example, where the optical sensing site is a well containing a sensor single stranded DNA ol igonucleotide having a fluorescent tag attached, a solution containing a target complementary single stranded DNA added to the well could biochemically interact by base-pairing with the sensor within the optical sensing site (not shown). In another example the optical sensing site is a location or well containing one or more immunoassay reagent for conducting an immunoassay as described herein.
[000308] In a particular embodiment, the optical sensing sites comprise optical transducers (not shown). An optical transducer is defined as any device that generates a measurable change (wavelength, amplitude or phase) to the incoming primary light wave which can thus be monitored in the outgoing secondary light wave. In one embodiment the optical transducers are fluorescence wells including fluorescent or luminescent compounds, wherein light waves guided by the waveguides excite the fluorescent or l uminescent compound in the wells in the presence of a target, and the same waveguides collect and guide light emitted from the wells to the detector (possibly through an adapter chip), for example at the edge of the chip (not shown).
[000309] The sensor of the optical sensing site of the system can be a sensor that discriminates or interacts with a target (e.g., a biologically active analyte) in a sample from, for example, a biological, man-made or environmental source. As discussed above, a first lightwave can induce the sensor to transduce an optical signal to a second light wave. In one embodiment where the sensor is capable of discriminating or interacting with a target in a sample, a measurable change in the second light wave can result when the sensor discriminates or interacts with the target. Upon detection of the change in the second light wave using the detector of the system, the presence of the target in the sample is indicated.
[000310J Any of a number of sensors can be used with the detection system to measure phenomena associated with the sensing of a target in a sample. Examples of suitable sensors include, but are not l imited to, a fluorescence well or cell, an absorption cell, an interferometric sensor, a diffractive sensor or a surface plasmon resonance (SPR) detector. For a fluorescence well or cell, the measurable phenomenon can be light emission from luminescent or fluorescent molecular tags. For example, emitted light at an altered wavelength can be measured. In the case of an absorption cell, changes in the sample optical density (OD) can measurably affect the intensity of the light passing through the sample. For an interferometric sensor, changes in the effective refractive index of a waveguide generate a phase difference between two light waves leading to different interference patterns measurable as a difference in i ntensity at the detector. For a diffractive sensor, changes in the effective refractive index at the surface of a diffract ive element, for example, a grating, affect the diffraction angle of the light for a given wavelength or alternatively affect the wavelength at a given diffraction angle. In the case of an SPR sensor, changes in the effect ive refractive index at a metal-dielectric interface affect the resonance conditions for generating surface plasmons. [00031 1 ] A control system for managing the different steps of operating the detection system is envisioned.
[000312] The control system can manage steps such as aligning and driving the scanning l ight source chip to scan the edge of the substrate, in addition to switching the light output from the light source, reading the detector array and reporting the results detected.
[000313] In general in one aspect, a method of using the systems and devices described herein to detect the presence of a single biologically active analyte molecule in a sample is provided. Biologically active analyte molecules in this context include any of the biologically active analyte molecules disclosed herein.
[000314] In practicing the methods of the present invention, many conventional techniques in molecular biology are optionally utilized. These techniques are well known and are explained in, for example, Ausubel et al. (Eds.) Current Protocols in Molecular Biology, Volumes I II, and III, ( 1997), Ausubel et al. (Eds.), Short Protocols in Molecular Biology: A Compendium of Methods from Current Protocols in Molecular Biology, 5th Ed., John Wiley & Sons, Inc. (2002), Sambrook et al., Molecular Cloning: A Laboratory Manual, 3rd Ed., Cold Spring Harbor Laboratory Press (2000), and Innis et al. (Eds.) PCR Protocols: A Guide to Methods and Applications, Elsevier Science & Technology
Books ( 1990), all of which are incorporated herein by reference.
[000315] Sample preparation suitable for use with the system and methods described herein can include any of a number of well known methods for collection and analysis of biological and/or environmental samples. In the case of biological samples the sample can be, for example, manipulated, treated, or extracted to any desired level of purity for a target of interest.
[000316] The sample can be a bodily fluid suspected of containing a biologically active analyte. Commonly employed bodily fluids include but are not l imited to blood, serum, saliva, urine, gastric and digestive fluid, tears, stool, semen, vaginal fluid, interstitial fluids derived from tumorous tissue, and cerebrospinal fluid.
[000317] It is anticipated that the systems described herein can be used for screening a large variety of samples. In the case where the investigated subject is a living creature, the sample may originate from body fluids as discussed. Methods of obtaining samples include but are not limited to cheek swabbing, nose swabbing, rectal swabbing, skin fat extraction or other collection strategies for obtaining a biological or chemical substance. When the tested subject is a non-living or environmental body, the sample may originate from any substance in a solid phase, liquid phase or gaseous phase. The sample may be col lected and placed onto the substrate or the substrate may be directly exposed to the investigated sample source (e.g. water reservoir, free air) and interact with it.
|000318] In some embodiments, the bodily fluids are used directly for detecting one or more biologically active analyte present therein without further processi ng. Where desired however, the bodily fl uids can be pre-treated before performing the analysis with the detection system. The choice of pre- Ireat ments wi l l depend on the type of bodily fluid used and/or the nature of the biologically active analyte under investigation. For instance, where the biological ly active analyte is present at a low level in a sample of bodily fluid, the sample can be concentrated via any conventional means to enrich the biologically active analyte. Methods of concentrating a biologically active analyte include but are not l imited to drying, evaporation, centrifugation, sedimentation, precipitation, and amplification. Where the biologically active analyte is a nucleic acid, it can be extracted using various lytic enzymes or chemical sol utions according to the procedures set forth in Sambrook et al . ("Molecular Cloning: A Laboratory Manual"), or using nucleic acid binding resins following the accompanying instructions provided by manufactures. Where the biological ly active analyte is a molecule present on or within a cell, extraction can be performed using lysing agents including but not limited to denaturing detergents such as SDS or nondenaturing detergents such as thesit (2-dodecoxyethanol), sodium deoxylate, Triton® X- 100, and Tween® 20.
[000319] In some embodiments, pretreatment can include diluting and/or mixing the sample, and filtering the sample to remove, e.g., red blood cells from a blood sample.
[000320] Targets detectable using the detection system include but are not limited to, a biologically active analyte including a nucleic acid, a protein, an antigen, an antibody, a microorganism, a gas, a chemical agent and a pollutant.
[000321] In one embodiment, the target is a nucleic acid that is DNA, for example, cDNA. In a related embodiment, the DNA target is produced via an amplification reaction, for example, by polymerase chain reaction (PCR). In another embodiment of the subject invention, the detected biologically active analyte is a protein representing a known biomarker for a disease or specific condition of the investigated organism. In another embodiment several different biologically active analytes can be proteins provided as a panel of bio-markers wherein relative concentrations of the bio-markers are indicative for a disease or other condition of the investigated organism. In a further embodiment the target is a microorganism that is a pathogen. In another embodiment the target is a chemical agent, for example, a toxic chemical agent.
[000322] Where the target is a nucleic acid, it can be single-stranded, double-stranded, or higher order, and can be linear or circular. Exemplary single-stranded target nucleic acids include mRNA,
I'RNA, tRNA, hnRNA, ssRNA or ssDNA virai genomes, although these nucleic acids may contain internally complementary sequences and significant secondary structure. Exemplary double-stranded target nucleic acids include genomic DNA, mitochondrial DNA, chloroplast DNA, dsRNA or dsDNA viral genomes, plasmids, phage, and viroids. The target nucleic acid can be prepared synthetically or purified from a biological source. The target nucleic acid may be purified to remove or diminish one or more undesired components of the sample or to concentrate the target nucleic acids. Conversely, where the target nucleic acid is too concentrated for the particular assay, the target nucleic acid may be di luted.
[000323] Fol lowing sample collection and optional nucleic acid extraction, the nucleic acid portion of the sample comprising the target nucleic acid can be subjected to one or more preparative reactions. These preparative reactions can include i n vitro transcription ( I VT), labeling, fragmentation, ampl ification and other reactions. mRNA can first be treated with reverse transcriptase and a primer to create cDNA prior to detection and/or ampl i fication; this can be done in vitro with puri fied mRNA or in situ, e.g. in cells or tissues affixed to a sl ide. Nucleic acid ampl ification increases the copy number of sequences of interest such as the target nucleic acid. A variety of amplification methods are suitable for use, including the polymerase chain reaction method (PCR), the ligase chain reaction (LCR), self sustained sequence replication (3SR), nucleic acid sequence-based amplification (NASBA), the use of Q Beta replicase, reverse transcription, nick translation, and the like.
[000324] Where the target nucleic acid is single-stranded, the first cycle of ampl ification forms a primer extension product complementary to the target nucleic acid. If the target nucleic acid is single stranded RNA, a polymerase with reverse transcriptase activity is used in the first amplification to reverse transcribe the RNA to DNA, and additional amplification cycles can be performed to copy the primer extension products. The primers for a PCR must, of course, be designed to hybridize to regions in their corresponding template that wi ll produce an amplifiable segment; thus, each primer must hybridize so that its 3 ' nucleotide is paired to a nucleotide in its complementary template strand that is located 3 ' from the 3' nucleotide of the primer used to replicate that complementary template strand in the PCR.
[000325] The target nucleic acid can be amplified by contacting one or more strands of the target nucleic acid with a primer and a polymerase having suitable activity to extend the primer and copy the target nucleic acid to produce a full length complementary nucleic acid or a smaller portion thereof. Any enzyme having a polymerase activity that can copy the target nucleic acid can be used, including DNA polymerases, RNA polymerases, reverse transcriptases, and enzymes having more than one type of polymerase activity, and the enzyme can be thermolabile or thermostable. Mixtures of enzymes can also be used. Exemplary enzymes include: DNA polymerases such as DNA Polymerase I ("Pol 1"), the
Klenow fragment of Pol I, T4, T7, Sequenase® T7, Sequenase® Version 2.0 T7, Tub, Tag, Tth, Pfx, Pfu, Tsp, Tfl, Tli and Pyrococcus sp GB D DNA polymerases; RNA polymerases such as E. coli, SP6, T3 and T7 RNA polymerases; and reverse transcriptases such as AMV, M MuLV, MMLV, RNAse H' MMLV (Superscript®), Superscript® I I, ThermoScript®, HI V 1 , and RAV2 reverse transcriptases. All of these enzymes are commercially available. Exemplary polymerases with multiple specificities include RAV2 and Tli (exo) polymerases. Exemplary thermostable polymerases include Tub, Taq, Tth, Pfx, Pfu, Tsp, Tfl, Tli mdPyrococcus sp. GB D DNA polymerases.
[000326] Suitable reaction conditions are chosen to permit amplification of the target nucleic acid, including pH, buffer, ionic strength, presence and concentration of one or more salts, presence and concentration of reactants and cofactors such as nucleotides and magnesium and/or other metal ions (e.g., manganese), optional cosolvents, temperature, and thermal cycling profi le for amplification schemes comprising a polymerase chain reaction, and may depend in part on the polymerase being used as well as the nature of the sample. Cosolvents include formamide (typically at from about 2 to about 10%).
glycerol (typical ly at from about 5 to about 10%), and DMSO (typically at from about 0.9 to about 1 0%). Techniques may be used in the amplification scheme in order to minimize the production of false positives or artifacts produced during amplification. These include ""touchdown" PCR, hot start techniques, use of nested primers, or designing PCR primers so that they form stem-loop structures in the event of primer-dimer formation and thus are not amplified. Techniques to accelerate PCR can be used, for example, centrifugal PCR, which al lows for greater convection within the sample, and comprising infrared heating steps for rapid heating and cooling of the sample. One or more cycles of amplification can be performed. An excess of one primer can be used to produce an excess of one primer extension product during PCR; preferably, the primer extension product produced in excess is the ampl ification product to be detected. A plurality of different primers may be used to amplify different target nucleic acids or different regions of a particular target nucleic acid within the sample.
[000327] Amplified target nucleic acids may be subjected to post amplification treatments. For example, in some cases, it may be desirable to fragment the target nucleic acid prior to hybridization in order to provide segments which are more readily accessible. Fragmentation of the nucleic acids can be carried out by any method producing fragments of a size useful in the assay being performed; suitable physical, chemical and enzymatic methods are known in the art.
[000328] An amplification reaction can be performed under conditions which allow a nucleic acid associated with the optical sensing site to hybridize to the ampl ification product during at least part of an amplification cycle. When the assay is performed in this manner, real time detection of this hybridization event can take place by monitoring for light emission during amplification.
[000329] Real time PCR product analysis (and related real time reverse-transcription PCR) provides a well-known technique for real time PCR monitoring that has been used in a variety of contexts, which can be adapted for use with the methods described herein (see, Laurendeau et al. ( 1999) "TaqMan PCR-based gene dosage assay for predictive testing in individuals from a cancer family with INK4 locus haploinsufficiency" Clin Chem 45(7):982-6; Bieche et al . ( 1999) "Quantitation of MYC gene expression in sporadic breast tumors with a real-time reverse transcription-PCR assay" Cancer
Res 59( 12):2759-65; and reuzer et al. ( 1999) "LightCycler technology for the quantitation of bcr/abl fusion transcripts" Cancer Res 59( 13):3 171 -4, all of which are incorporated by reference). In addition, l inear PCR and Linear-After-The Exponential (LATE)-PCR can be adapted for use with the methods described herein.
[000330] Immunoassays can be conducted on the detection system of the invention, for example, at one or more optical sensing site of the system. Suitable immunoassay systems include but are not limited to competitive and noncompetitive assay systems. Such assay systems are typically used with techniques such as western blots, radioimmunoassays, EIA (enzyme immunoassay), ELISA (enzyme-linked immunosorbent assay), "sandwich" immunoassays, immunoprecipitation assays, precipitin reactions, gel diffusion precipitin reactions, immunodiffusion assays, agglutination assays, complement-fixation assays, immunoradiometric assays, fluorescent immunoassays, protein A immunoassays, and cellular immunostaining (fixed or native) assays to name but a few. Such assays are routine and well known in the art (see, e.g., Ausubel et al ., supra). Immunoassay techniques particularly useful with the detection systems described herein include but are not limited to ELISA, "sandwich" immunoassays, and fluorescent immunoassays. Exemplary immunoassays are described briefly below (but are not intended by way of l imitation). [000331 ] ELISAs generally involve preparing antigen, coating a wel l (e.g., an optical sensing site of the detection system) with the antigen, adding the antibody of interest conjugated to a detectable compound such as an enzymatic substrate (e.g., horseradish peroxidase or alkaline phosphatase) to the well and incubating for a period of time, and detecting the presence of the antigen. In ELISAs the antibody of interest does not have to be conjugated to a detectable compound; instead, a second antibody (which recognizes the antibody of interest) conjugated to a detectable compound may be added to the well. Further, instead of coating the well with the antigen, the antibody may be coated to the well. In this case, a second antibody conjugated to a detectable compound may be added following the addition of the antigen of interest to the coated well. One of ski ll in the art would be knowledgeable as to the parameters that can be modified to increase the signal detected as well as other variations of ELISAs known in the art.
[000332] In one exemplary immunoassay, a sample contains an unknown amount of biologically active analyte to be measured, which may be, for example, a protein. The analyte may also be termed an antigen. The sample may be spiked with a known or fixed amount of labeled analyte. The spiked sample is then incubated with an antibody that binds to the analyte, so that the analyte in the sample and the labeled analyte added to the sample compete for binding to the available antibody binding sites. More or less of the labeled analyte will be able to bind to the antibody binding sites, depending on the relative concentration of the unlabeled analyte present in the sample. Accordingly, when the amount of labeled analyte bound to the antibody is measured, it is inversely proportional to the amount of unlabeled analyte in the sample. The amount of analyte in the original sample may then be calculated based on the amount of labeled analyte measured, using standard techniques in the art.
[000333] In one exemplary competitive immunoassay, an antibody that binds to a biologically active analyte may be coupled with or conjugated with a ligand, wherein the ligand binds to an additional antibody added to the sample being tested. One example of such a ligand includes fluorescein. The additional antibody may be bound to a solid support (e.g., an optical sensing site of the detection system). The additional antibody binds to the ligand coupled with the antibody that binds in turn to the analyte or alternatively to the labeled analyte, forming a mass complex which allows isolation and measurement of the signal generated by the label coupled with the labeled analyte.
[000334] In another type of exemplary competitive immunoassay, the biologically active analyte to be measured may be bound to a solid support (e.g., an optical sensing site of the detection system), and incubated with both an antibody that binds to the analyte and a sample containing the analyte to be measured. The antibody binds to either the analyte bound to the solid support or to the analyte in the sample, in relative proportions depending on the concentration of the analyte in the sample. The antibody that binds to the analyte bound to the solid support is then bound to another antibody, such as anti-mouse IgG, that is coupled to a label . The amount of signal generated from the label is then detected to measure the amount of antibody that bound to the analyte bound to the sol id support. Such a measurement will be inversely proportional to the amount of analyte present in the sample. Such an assay may be used in the detection system of the present invention. |000335] A wide diversity of labels are available in the art that can be employed for conducting the subject assays. In some embodiments labels are detectable by spectroscopic, photochemical, biochemical, immunochemical, or chemical means. For example, useful nucleic acid labels include fluorescent dyes, enzymes, biotin, dioxigenin, or haptens and proteins for which antisera or monoclonal antibodies are avai lable. A wide variety of labels suitable for labeling biological components are known and are reported extensively in both the scientific and patent literature, and are generally applicable to the present invention for the labeling of biological components. Suitable labels include enzymes, substrates, cofactors, inhibitors, fluorescent moieties, chemi luminescent moieties, or bioluminescent labels. Label ing agents optionally include, for example, monoclonal antibodies, polyclonal antibodies, proteins, or other polymers such as affinity matrices, carbohydrates or lipids. Detection proceeds by any of the methods described herein, for example, by detecting an optical signal in an optical waveguide. A detectable moiety can be of any material having a detectable physical or chemical property. Such detectable labels have been well-developed in the field of gel electrophoresis, column chromatography, solid substrates, spectroscopic techniques, and the like, and in general, labels useful in such methods can be applied to the present invention. Preferred labels include labels that produce an optical signal. Thus, a label includes without limitation any composition detectable by spectroscopic, photochemical, biochemical, immunochemical, electrical, optical, thermal, or chemical means.
[000336] In some embodiments the label is coupled directly or indirectly to a molecule to be detected such as a product, substrate, or enzyme, according to methods well known in the art. As indicated above, a wide variety of labels are used, with the choice of label depending on the sensitivity required, ease of conjugation of the compound, stability requirements, available instrumentation, and disposal provisions. Non radioactive labels are often attached by indirect means. Generally, a ligand molecule is covalently bound to a polymer. The ligand then binds to an anti ligand molecule which is either inherently detectable or covalently bound to a signal system, such as a detectable enzyme, a fluorescent compound, or a chemiluminescent compound. A number of ligands and anti-ligands can be used. Where a ligand has a natural anti-ligand, for example, biotin, thyroxine, and Cortisol, it can be used in conjunction with labeled anti-ligands. Alternatively, any haptenic or antigenic compound can be used in combination with an antibody.
[000337] In some embodiments the label can also be conjugated directly to signal generating compounds, for example, by conjugation with an enzyme or fluorophore. Enzymes of interest as labels will primarily be hydrolases, particularly phosphatases, esterases and glycosidases, or oxidoreductases, particularly peroxidases. Fluorescent compounds include fluorescein and its derivatives, rhodamine and its derivatives, dansyl, and umbell iferone. Chemi luminescent compounds include luciferin, and 2,3- dihydrophthalazinediones, such as luminol .
[000338] Methods of detecting labels are wel l known to those of ski l l in the art. Thus, for example, where the label is a fluorescent label, it may be detected by exciting the tluoi chrome with the appropriate wavelength of light and detecting the resulting fluorescence by. for example, a detection system as described herein. Simi larly, enzymatic labels are detected by providing appropriate substrates for the enzyme and detecting the resulting reaction product (e.g., a reaction product capable of producing a detectable optical signal).
[000339] In some embodiments the detectable signal may be provided by luminescent sources. "Luminescence" is the term commonly used to refer to the emission of light from a substance for any reason other than a rise in its temperature. In general, atoms or molecules emit photons of electromagnetic energy (e.g., light) when they move from an "excited state" to a lower energy state (usually the ground state); this process is often referred to as "radioactive decay". There are many causes of excitation. If the exciting cause is a photon, the luminescence process is referred to as "photoluminescence". If the exciting cause is an electron, the luminescence process is referred to as "electroluminescence". More specifically, electroluminescence results from the direct injection and removal of electrons to form an electron-hole pair, and subsequent recombination of the electron-hole pair to emit a photon. Luminescence which results from a chemical reaction is usually referred to as "chemiluminescence". Luminescence produced by a living organism is usually referred to as "bioluminescence". If photoluminescence is the result of a spin allowed transition (e.g., a single-singlet transition, triplet-triplet transition), the photoluminescence process is usually referred to as "fluorescence". Typically, fluorescence emissions do not persist after the exciting cause is removed as a result of short-lived excited states which may rapidly relax through such spin allowed transitions. If photoluminescence is the result of a spin forbidden transition (e.g., a triplet- singlet transition), the photoluminescence process is usually referred to as "phosphorescence". Typically, phosphorescence emissions persist long after the exciting cause is removed as a result of long-lived excited states which may relax only through such spin-forbidden transitions. A "luminescent label" may have any one of the above-described properties.
[000340] Suitable chemiluminescent sources include a compound which becomes electronically excited by a chemical reaction and may then emit light which serves as the detectible signal or donates energy to a fluorescent acceptor. A diverse number of families of compounds have been found to provide chemiluminescence under a variety of conditions. One family of compounds is 2,3-dihydro- l ,4- phthalazinedione. A frequently used compound is luminol, which is a 5-amino compound. Other members of the family include the 5-amino-6,7,8-trimethoxy- and the dimethylamino[ca]benz analog. These compounds can be made to luminesce with alkaline hydrogen peroxide or calcium hypochlorite and base. Another family of compounds is the 2,4,5-triphenyl imidazoles, with lophine as the common name for the parent product. Chemi luminescent analogs include para-dimethyl ami no and -methoxy substituents. Chemiluminescence may also be obtained with oxalates, usually oxalyl active esters, for example, p-nitrophenyl and a peroxide such as hydrogen peroxide, under basic conditions. Other useful chemiluminescent compounds that are also known include— -alkyl acridinum esters and dioxetanes. Alternatively, luciferins may be used in conj unction with luciferase or lucigenins to provide
bioluminescence.
[000341 ] In a separate embodiment, the present invention provides a method of monitoring one or more pharmacological parameter, for example, pharmacodynamic (PD) and/or pharmacokinetic (PK) parameters, useful for assessing efficacy and/or toxicity of a therapeutic agent. The method comprises subjecting a sample of bodily fluid from a subject administered with the therapeutic agent to a detection device for monitoring the one or more pharmacological parameter; using the detection device as described herein to yield detectable signals indicative of the values of the more than one pharmacological parameter from the sample; and detecting the detectable signal generated from said sample of bodily fluid.
[000342] In one implementation the samples tested can include a large number of a variety of small molecules (e.g., screening libraries) which are of interest when investigating new drugs. Accordingly, the detection system described herein is useful for screening libraries of small molecules to investigate their ability to interact with certain biologically active analytes to reveal potential new drugs. Further screening of some or all small molecule candidates may reveal adverse drug effects and toxicity.
[000343] In one implementation the samples can include molecules which are tested for toxicity.
[000344] In general in another aspect methods of using the detection systems described herein are provided.
[000345] In one embodiment, the scanning light source moves through its scanning path to a point at which it is coupled to and in optical communication with one or more in-coupling or excitation waveguides, thus generating a pulse of light within the waveguides. The light travels along the waveguides, reaches the optical sensing sites and interacts through the sensor, for example, an optical transducer. The samples are positioned at or near the waveguides. Next, the secondary light leaving the sensor couples into the out-coupling or collection waveguides and travels down the waveguide to its end at an edge of the substrate, for example, a chip facet. Light exiting the out-coupling or collection waveguides is then detected by the different elements of the detector, which can be a detector array. In some embodiments, the substrate comprises a plurality of waveguides that serve as both in/out-coupling waveguides, with the light source and the detector being coupled to and in optical communication with opposite ends of the waveguides. In other embodiments, the in/out-coupl ing waveguides collect light from the light source and guide the secondary light to the detector through one or more adapter.
[000346] In another embodiment, the scanning light source/detector moves through its scanning path to a point at which the light source is coupled to and in optical communication with one or more in- coupling waveguides, thus generating a pulse of light within the waveguides. At the same time, the detector is coupled to and in optical communication with one or more out-coupling waveguides. The l ight travels along the waveguides, reaches the optical sensing sites and interacts through the sensor, for example, an optical transducer. The samples are positioned at or near the waveguides. Next, the secondary light leaving the sensor couples into the out-coupling waveguides and travels down the waveguide to its end at an edge of the substrate, for example, a chip facet. Light exiting the out-coupl ing waveguides is then detected by the different elements of the detector, which can be a detector array. In some embodiments, the substrate comprises a plural ity of waveguides that serve as both in/out-coupling waveguides, while the light source/detector chip comprises in-coupling and out-coupling waveguides that are coupled through at least one combiner. The light waves generated by the light source elements are coupled into the in-coupl ing waveguides of the l ight source/detector chip, then combined by combiners into the out-coupling waveguides, which couple this primary light to the waveguides of the substrate. The secondary l ight wave leaving the sensor travels through the same waveguides of the substrate to out- coupling waveguides of the light source/detector chip which guide the light wave to the detector elements.
[000347] In another embodiment, detection of a sample includes delivering a sample suspected of containing a target to be detected to an optical sensing site of the detection system. Delivering a sample to the system can include pipetting of a fluid to the optical sensing site. Other delivery means can include but are not limited to a robotic fluid delivery system or physically depositing a non-fluid or semi-fluid sample at the optical sensing site, either by hand or with the aid of a tool or robot manipulation system. Next, a first light wave produced by the scanning light source is provided to one or more of the plurality of waveguides in optical communication with the optical sensing site. The first light wave is transduced (e.g., measurably changed) by the sensor associated with the optical sensing site to form a second light wave carried back in one or more of the plurality of out-coupling or collection waveguides which are in optical communication with the optical sensing site. Next a measurable change in the second light wave is detected using the detector which is in optical communication with the out-coupling or col lection waveguides. Detection of a measurable change in the second light wave indicates that the sensor has interacted with the target. It is envisioned that in various embodiments the waveguides described herein can be arranged substantially parallel as illustrated general ly in the accompanying figures.
[000348] In a further embodiment, the detection method includes generating one or more light wave by the scanning light source which couples into the substrate at some point along its scanning path to produce the first light wave in one or more of the waveguides in a controlled manner.
[000349] In another embodiment, the different light source elements of the scanning light source can be switched on simultaneously to generate one or more input light waves. The plural ity of light waves can be coupled into the substrate to controllably produce the first light wave in one or more of the waveguides.
[000350] In one embodiment, all in-coupling waveguides are provided with a first light wave and simultaneous detection of second light waves at each out-coupling waveguide is achieved using a detector that is a photodetector array.
[000351] By controlled switching of the different light source elements, each waveguide can be individually addressed with a first light wave. The order of addressing the waveguides can be sequential, staggered, random or in any order desired. Rapid scanning of the entire array of optical sensing sites can be achieved with the aid of the photodetector array since any second light wave associated with each out- coupling waveguide can be simultaneously detected.
[000352] In another embodiment, a single excitation waveguide is provided with a first l ight wave and simultaneous detection of second l ight waves at each col lection waveguide is achieved using a detector that is a photodetector array. For example, where the two-dimensional waveguide array is configured as an array of 128 excitation waveguides and 128 collection waveguides, then it would be possible to simultaneously detect second light waves (if any) generated from 1 28 optical sensor sites after providing a single first lightwave in a first excitation waveguide. Thus, 128 optical sensing sites can be interrogated for presence or absence of target simultaneously. Next, a second excitation waveguide can be provided thereby triggering the interrogation of a second set of 128 optical sensing sites. The process can rapidly be repeated until every excitation waveguide has been excited and the entire array of optical sensing sites have been interrogated.
[000353] In various embodiments the method of using the detection system involves the detection of a substance, including but not limited to a biologically active analyte including a nucleic acid, a protein, an antigen, an antibody, a panel of proteins, a microorganism, a gas, a chemical agent and a pollutant. In a particular embodiment, a single nucleotide polymorphism (SNP) is detected in the target. In one embodiment expression of a gene is detected upon detection of the target.
[000354] Systems using planar waveguides for optical detection of SNPs have been described before. For example, single base extension ("SBEX") with planar waveguide fluorescent biosensor technology to detect SNPs has been described by Herron and Tolley in U.S. patent application Ser. No. 10/984,629, filed Nov. 8, 2004 and titled "Single Base Extension." Briefly, total internal reflectance fluorometry (TIRF) can be used in combination with SBEX under real time detection conditions for SNP detection using planar waveguide technology. Evanescent waves generated in a waveguide substrate wi ll only excite fluorescently labeled analyte DNA molecules that are bound to stationary capture oligonucleotides. Herron found that the depth of evanescent wave useful for measurements is within about 300 nm of the sensor surface. The SBEX approach uses a DNA polymerase to incorporate, for example, Cy5 labeled dideoxynucleotriphosphates (ddNTPs). Additional labels are discussed elsewhere herein.
[000355] Identification ("call ing") of the single base added to the 3' end of the probe molecule can be done in one of three ways: parallel channels for each of the four bases using a different labeled ddNTP in each channel ; sequential SBEX reactions using a different labeled ddNTP in each reaction; or wavelength discrimination of the four possibil ities using a different fluorescent label for each ddNTP. The first of these methods may be preferred. SBEX may be used in oligonucleotide genotyping and SNP detection systems, and is advantageous over traditional hybridization assays, for example, due to greater base specificity, production of a covalent bond between the labeled ddNTP and the probe, and simultaneous detection of multiple bases.
[000356] By using SBEX on waveguides, simultaneous detection of several different
polymorphisms can be done with ease. By patterning the waveguide with different capture sequences, different points in a sequence, for example, a genome, a chromosome and/or a gene, may be assayed. As SBEX only requires a fluorescent label on the ddNTP monomers used, all instances of a particular base wi ll be detected. In order to do the same thing with a traditional DNA hybridization assay, each probe DNA for each capture sequence would have to be fluorescently labeled.
|000357] The enzyme-catalyzed reaction has two distinct advantages. First, a stable covalent bond forms between the stationary phase and a labeled monomer, e.g., a Cy5-labeled monomer. This increases the assay sensitivity versus traditional hybridization assays where the fluorescent label is captured by the stationary phase via non-covalent interactions (duplex formation). Optionally a stringent washing step can be employed. Second, the polymerase enzyme incorporates the dideoxynucleotide with high fidelit— due to the replication accuracy of a polymerase, in general only the base that is complementary to the target base will react. SBEX is particularly well suited for planar waveguide technology, benefiting from the increased speed of a washless assay and increased sensitivity provided by kinetic data.
[000358] Using SBEX on the waveguide platform enables rapid assays (<5 min) to be performed that are able to differentiate between single nucleotide polymorphic and wild type sequences at temperatures less than 50° C.
[000359] Fluorescence imaging is sensitive to speed, sensitivity, noise and resolution, and each may be optimized for use in the invention; for example, speed may be increased to decrease assay times. Base extension may be detected using a CCD camera, a streak camera, spectrofluorometers, fluorescence scanners, or other known fluorescence detection devices, which generally comprise four elements: an excitation source; a fluorophore; a filter to separate emission and excitation photons; and a detector to register emission photons and produce a recordable output, typically an electrical or photographic output.
[000360] Polymerase enzymes useful in the invention are known in the art and include, but are not limited to, thermostable polymerases, such as pfu, Taq, Bst, Tfl, Tgo and Tth polymerase, DNA
Polymerase I, Klenow fragment, and/or T4 DNA Polymerase. The polymerase may be a DNA-dependent DNA polymerase, a DNA-dependent RNA polymerase, a RNA-dependent RNA polymerase, a RNA- dependent DNA polymerase or a mixture thereof, depending on the template, primer and NTP used. The polymerase may or may not have proofreading activity (3 ' exonuclease activity) and/or 5' exonuclease activity).
[000361] The capture molecule and/or the analyte molecule of the invention may be any nucleic acid, including, but not limited to, DNA and/or RNA and modifications thereto known in the art, and may incorporate 5'-0-( l -thio)nucleoside analog triphosphates, . alpha. -thiotriphosphate, 7-Deaza-.alpha.- thiotriphosphate, N6-Me-.alpha. -thiotriphosphate, 2'-0-Methyl-triphosphates, morpholino, PNA, aminoalkyl analogs, and/or phosphorothioate.
[000362] In one embodiment immunoassays can be used with the present method of using the detection system. The optical sensing site of the detection system of the invention can be adapted to support an immunoassay, for example, by including one or more immunoassay reagent at or withi n the optical sensing site. In this embodiment an interaction between the optical sensing site and a sample being tested for a biologically active analyte can include an immunoassay conducted at the optical sensing site. As such, the optical sensing site interacting with the biologically active analyte can include an outcome of an immunoassay. In this manner, presence or absence of the analyte can be determined. Additional ly the amount of analyte can be quantified. In one embodiment the immunoassay supported is a fluorescent assay. It is envisioned that the immunoassay can be a competitive or non-competitive immunoassay. In one embodiment the immunoassay supported is an EL1SA.
[000363] It is envisioned thai a variety of instrumentation relating to biological or environmental sample preparation, handling and analysis can be used in conjunction with the system and methods described herein. Examples of such instrumentation include but are not l imited to a cell sorter, a DNA ampl ification thermal cycler, or a chromatography machine (e.g., GC or HPLC). Such instrumentation is wel l known to those skil led in the art. It is envisioned that a robotic interface could be used between the detection system of the present invention and various instrumentation relating to biological or environmental sample preparation, handling and analysis.
[000364] The optical detection system may be used in a range of applications including biomedical and genetic research as well as clinical diagnostics. Arrays of polymers such as nucleic acids may be screened for specific binding to a target, such as a complementary nucleotide, for example, in screening studies for determination of binding affinity and in diagnostic assays. In one embodiment, sequencing of polynucleotides can be conducted, as disclosed in U.S. Pat. No. 5,547,839. The nucleic acid arrays may be used in many other appl ications including detection of genetic diseases such as cystic fibrosis or diagnosis of diseases such as HIV, as disclosed in U.S. Pat. No. 6,027,880 and U.S. Pat. No. 5,861 ,242. Genetic mutations may be detected by sequencing or by hydridization. In one embodiment, genetic markers may be sequenced and mapped using Type-IIs restriction endonucleases as disclosed in U.S. Pat. No. 5,710,000.
[000365] Other applications include chip based genotyping, species identification and phenotypic characterization, as described in U.S. Pat. No. 6,228,575. Still other applications including diagnosing a cancerous condition or diagnosing viral, bacterial, and other pathological or nonpathological infections, are described in U.S. Pat. No. 5,800,992. A further application includes chip based single nucleotide polymorphism (SNP) detection as described in U.S. Pat. No. 6,361 ,947.
[000366] Gene expression may be monitored by hybridization of large numbers of mRNAs in parallel using high density arrays of nucleic acids in cells, such as in microorganisms such as yeast, as described in Lockhart et a! ., Nature Biotechnology, 14: 1675-1 680 ( 1996). Bacterial transcript imaging by hybridization of total RNA to nucleic acid arrays may be conducted as described in Saizieu et al., Nature Biotechnology, 16:45-48 ( 1998). Accessing genetic information using high density DNA arrays is further described in Chee, Science 274:610-614 ( 1996).
[000367] In addition to the nucleic acid arrays discussed above, optical detection systems of the invention may be used in combination with protein and chemical microarrays, including arrays of protei ns, antibodies, small molecule compounds, peptides, and carbohydrates, or cell or tissue arrays, as described for example in Xu, Q. and Lam, K. S., J. Biomed. Biotechnol. 5:257-266 (2003). Protein and chemical arrays may be used in combination with the methods and devices of the invention in a number of potential applications, including but not l imited to proteomics (including assays of both protein-protein interactions and protein-l igand interactions), screening assays for drug discovery, and toxicology testing. In some appl ications these assays may uti l ize label-free optical sensing methods as described, for example, in U.S. Pat. No. 7,349,080 and U.S. Pat. No. 7,292,336.
[000368] A further potential application is detection of chemical and/or biological warfare agents, including but not limited to bacterial spores (for example, as descri bed in U .S. Pat. No. 6,498.04 1 ): bacterial agents (e.g., Bacillus anthracis, Yersinia pest is, F. titlararensis, Bruce/la, Clostridium Botulinum, Clostridium tetani, Coxiella burnetii, and Vibrio cholerae); viral agents (e.g., variola virus, viral encephalitis agents such as Venezuelan equine encephalitis, western equine encephal itis and eastern equine encephalitis; and viral hemorraghic fever agents such as arenaviridae, bunyaviridae, filoviridae, and flaviviridae) and toxins (e.g., Staphylococcus enterotoxin B, botuhnum toxin, ricin, and mycotoxins, as well as anticrop agents (e.g., Pitccinia graministrititi, Piricularia oryzae, Tileltia caries, Tilettia foetida, Fusarium fungus, and herbicides). Bacterial and viral agents may be detected using, for example, nucleic acid based methods such as real-time PCR, antibody-based detection methods such as ELISA, or using antimicrobial peptides as described by Kulagina et al., Sens. Actuators B. Chem. 121 : 1 50- ] 57 (2007).
[000369] Further potential applications are in food safety, including detection of food-borne pathogens (e.g., Salmonella typhosa, Salmonella typhimurium, Campylobacter jejuni, Escherichia coli 0157H:H7, Listeria monocytogenes, Stapholococcus aureus, and Clostridium perfringens); detection of chemical substances that function as indicators of deterioration, for example, those caused by the process of oxidation; and detection of traces of contaminating chemical compounds, toxins, additives, or pesticides.
[000370] Further non-limiting potential applications include detection and diagnosis of viral and bacterial infectious diseases (e.g. AIDS, Bird Flu, SARS, West Nile virus); point-of-care monitoring of patients (e.g, detection of sugar and insul in levels in diabetic patients, detection of blood gas levels or lactic acid levels); pregnancy testing; detection of drugs or narcotics (e.g., cocaine, ecstasy,
methamphetamines, opiates); detection of chemical and explosive substances (e.g, RDX, TNT, nitroglycerin); and environmental monitoring of air, water or soil samples, including the detection of pesticides, heavy metals, nitrates or phosphates.
[000371 ] The working system described here can also be a sub-system within a much larger bio- analysis system. The bio-analysis system could include all the aspects of sample preparation prior to optical detection, the post processing of data collected in the optical detection phase and finally decision making based on these results. Sample preparation may include steps such as: extraction of the sample from the tested subject (human, animal, plant environment etc.); separation of different parts of the sample to achieve higher concentration and purity of the molecules under investigation; sample amplification (e.g. through PCR); attachment of fluorescence tags or markers to different parts of the sample; and spotting of the sample onto the substrate. The post processing of the collected data may include: normalization; background and noise reduction; and statistical analysis such as averaging over repeated tests or correlation between different tests. The decision making may include: testing against a predefined set of rules and comparison to information stored in external databases.
[000372] The applications and uses of the detection systems described herein can produce one or more result useful to diagnose a disease state of an individual, for example, a patient. I n one embodiment, a method of diagnosing a disease comprises reviewing or analyzing data relating to the presence and/or the concentration level of a target i n a sample. A concl usion based upon review or analysis of the data can be provided to a patient, a health care provider or a health care manager. I n one embodiment the conclusion is based upon the review or analysis of data regarding a disease diagnosis. It is envisioned in another embodiment that providing a conclusion to a patient, a health care provider or a health care manager includes transmission of the data over a network.
[000373J Accordingly, business systems and methods using the detection systems and methods described herein are provided.
[000374] One aspect of the invention is a business method comprising screening patient test samples for the presence or absence of a biological ly active analyte to produce data regarding the analyte, collecting the analyte data, and providing the analyte data to a patient, a health care provider or a health care manager for making a conclusion based upon review or analysis of the data regarding a disease diagnosis. In one embodiment providing a conclusion to a patient, a health care provider or a health care manager includes transmission of the data over a network.
[000375] Accordingly FIG. 15 is a block diagram showing a representative example of a logic device through which reviewing or analyzing data relating to the present invention can be achieved. Such data can be in relation to a disease, disorder or condition in an individual. FIG. 15 shows a computer system (or digital device) 1500 connected to an apparatus 1 520 for use with the detection system 1524 to, for example, produce a result. The computer system 1 500 may be understood as a logical apparatus that can read instructions from media 151 1 and/or network port 1505, which can optional ly be connected to server 1 509 having fixed media 1 512. The system shown in FIG. 1 5 includes CPU 1 501 , disk drives 1503, optional input devices such as keyboard 1515 and/or mouse 1 516 and optional monitor 1507. Data communication can be achieved through the indicated communication medium to a server 1 509 at a local or a remote location. The communication medium can incl ude any means of transmitting and/or receiving data. For example, the communication medium can be a network connection, a wireless connection or an internet connection. Such a connection can provide for communication over the World Wide Web. It is envisioned that data relating to the present invention can be transmitted over such networks or connections for reception and/or review by a party 1 522. The receiving party 1 522 can be but is not limited to a patient, a health care provider or a health care manager.
[000376] In one embodiment, a computer-readable medium includes a medium suitable for transmission of a result of an analysis of an environmental or biological sample. The medium can include a result regarding a disease condition or state of a subject, wherein such a result is derived using the methods described herein.
|000377] K its comprising reagents useful for performing the methods described herein are also provided.
[000378] In some embodiments, a kit comprises detection system as described herein and reagents for detecti ng a target in the sample. The kit may optionally contain one or more of the fol lowing: one or more fluorescent or l uminescent molecular tag, and one or more biologically active analyte incl uding a nucleic acid, protein, microorganism or chemical agent.
[000379] The components of a kit can be retained by a housing. Instructions for using the kit to perform a described method can be provided with the housing, and can be provided in any fixed medium. The instructions may be located inside the housing or outside the housing, and may be printed on the interior or exterior of any surface forming the housing that renders the instructions legible. A kit may be in multiplex form for detection of one or more different target biologically active analytc including nucleic acid, protein, microorganism, gas, chemical agent or pollutant.
[000380] As described herein and shown in an illustrative examples in FIG. 16, in certain embodiments a kit 1603 can include a detection system 1600, a housing or container 1 602 for housing various components. As shown in FIG. 16, and described herein, the kit 1603 can optionally include instructions 1601 and reagents 1 605, for example, DNA hybridization or immunoassay reagents. Other embodiments of the kit 1603 are envisioned wherein the components include various additional features described herein.
[000381 ] In one embodiment, a kit for assaying a sample for a target includes a detection system including a scanning light source, a detector, and a substrate. The substrate can include a plural ity of excitation waveguides and a plurality of collection waveguides as described herein. The excitation waveguides and collection waveguides of the substrate cross or intersect to form intersection regions and a two-dimensional array. The system further includes a plural ity of optical sensing sites. The optical sensing sites are in optical communication with one or more excitation waveguides and one or more collection waveguides. The kit further includes packaging and instructions for use of the system.
[000382] In another embodiment, the crossing of the excitation waveguides and collection waveguides is substantially perpendicular.
[000383] In another embodiment, a kit for assaying a sample for a target includes a detection system including a scanning light source, a detector, and a substrate. The substrate can include a plurality of substantially parallel in-coupling waveguides and a plurality of substantially parallel out-coupling waveguides as described herein. The system can further include a plurality of optical sensing sites. The optical sensing sites are in optical communication with one or more waveguides. The kit further includes packaging and instructions for use of the system.
[000384] In one embodiment, the kit includes a detection system that is a planar l ightwave circuit (PLC).
[000385] In general, in another aspect methods of manufacturing a substrate for assaying a sample for a target are provided. In one embodiment the substrate is a PLC.
[000386] The starting material for manufacturing PLC devices is a wafer usual ly made of silicon (Si) or sil ica (Si02). The most common wafer diameters in use are 4". 6" and 8". The manufacturing process for PLC devices involves two basic processes, namely, deposition and etching. A short description of each of them is given below.
[000387] In certain embodiments the methods of manufacturing the systems described herein can include, but are not limited to laser writing, UV writing and photonic band-gap waveguide methods. The manufacturing process in some embodiments includes one or more steps of deposition, masking and etching.
1000388] Deposition: [000389] In the deposition step a layer of well defined material having well controlled thickness is deposited across the entire wafer. The most common materials used for waveguide layer deposition are silica (Si02), also known as glass, and silicon nitride (Si3N4). The optical properties of the silica (mainly its refractive index) is control led by the amount of doping (Ge, P, and B etc.) introduced during the deposition. Other materials such as silicon, glass, epoxy, lithium niobate, indium phosphide and SiON (silicon oxynitride) and its derivatives are also used. For the cladding layer, materials can include but are not limited to si licon, si lica (Si02), glass, epoxy, lithi um niobate and indium phosphide.
[000390] The deposition step is done using several technologies such as PECVD (Plasma- Enhanced Chemical Vapor Deposition), LPCVD (Low Pressure CVD), APCVD (Atmospheric pressure CVD), FHD (Flame Hydrolysis Deposition) and others well known in the art.
[000391] FIG. 17A illustrates an exemplary substrate 1 704 as a schematic structure created after two consecutive deposition steps of a cladding 1721 layer and a core 1723 layer over a silicon 1726 layer, which can be a wafer. As mentioned above, these two layers differ in the refraction index which is achieved by using different levels of doping. Typical thicknesses for the different layers are: Cladding up to about 20 μιη and core up to 6 μηι. The thickness of the silicon 1 726 wafer can range from about 0.5 mm to 1 mm.
[000392] Masking:
[000393] Fol lowing the deposition and before the etching step, the desired two-dimensional structure of the PLC device is transferred to the deposited wafer by masking the areas not to be etched away. The masking is done in several steps involving covering the wafer with light sensitive material, exposing it to light through l ithographic masks and removing the exposed material leaving in place the mask. The result of such steps is shown in FIG. 17B where a mask 1 725 is shown on top of the core 1723 layer of the substrate 1704.
[000394] Etching:
[000395] In the etching step, material at the un-masked areas is removed from the top
core 1723 layer of the substrate (see FIG. 17C). The etching rate is a known parameter, therefore the etching depth can be control led by time. The two most common techniques for etching are wet-etching and Reactive-Ion-Etching (RIO). FIG. 17C shows the results of the etching step which results in a waveguide 1 727.
[000396] After the etching step, an over-cladding or top cladding 1 729 layer is created using a deposition step similar to the one described above. The results are shown in FIG. 1 7D. As shown in FIG. 1 7D, the resulting waveguide 1727 can be surrounded by a top cladding 1 729 and a cladding 1721 over a si licon 1726 layer.
[000397] The above steps can be repeated to create several waveguide layers one on top of the other. In this case, a planarization step may be required between one waveguide layer and the other. This is done using a technique known as Chemical Mechanical Planarization (CM P).
[000398] When the wafer processing is completed, it can be diced into the individual chips. An exemplary simpl ified flow-chart of the manufacturing process is shown in FIG. I 8. [000399J While preferred embodiments of the present invention have been shown and described herein, it will be obvious to those ski lled in the art that such embodiments are provided by way of example only. Numerous variations, changes, and substitutions will now occur to those skilled in the art without departing from the invention. It should be understood that various alternatives to the
embodiments of the invention described herein may be employed in practicing the invention. It is intended that the following claims define the scope of the invention and that methods and structures within the scope of these claims and their equivalents be covered thereby.
[000400] Signal Normal ization
[000401] The present invention features methods and optical arrangements enabling the performance of two or more assays separated by time and / or separated by the color / wavelength of their emitted fluorescence on each and every sensing-well of a waveguide based bio-sensing chip. In some embodiments, at least one of the assays performed in each and every well is attempting to measure the signal generated by the presence of a target moiety. The other assays measure the signal generated by one or more secondary moieties in the sample, either naturally occurring or intentionally added, or by moieties immobil ized to the sensing wel l, which signal indicates the level of a critical parameter such as temperature, excitation-light intensity, reagents aging, blood hematocrit and non-specific binding. These signals generated by the secondary moieties can serve to calibrate and / or normalize the signal measured from the target moiety in order to achieve a more consistent and reliable assay result which takes into account the variation of these signal affecting parameters.
[000402] The optical arrangement that can be used for the current invention may be based on the optical system as described above and further described in US 7,951 ,583 and US 8, 1 87,866 and can include in addition:
[000403] Two or more excitation lasers, each having a different wavelength.
[000404] Combining and switching mechanisms for getting the l ight from each laser to every sensing wel l on the chip.
[000405] Interference filters allowing one or more of the fluorescent signals excited by one or more of the lasers to reach the optical detector at the output of the sensing chip while blocking all excitation light from the lasers.
[000406] Moieties labelled with different fluorescent tags which fluorescence can be excited using one or more of the excitation lasers and can be measured through one or more of the interference filters.
[000407] In general, one or more specific capture moiet ies (e.g. antibodies specific to a target molecules) are immobilized in each of the sensing wel ls of the waveguide-based sensing chip. When the sample is introduced to the surface of the chip, the specific target moieties i n the sample start binding to the corresponding specific capture moieties in the sensing well. Shinning laser light having a wavelength within the excitation spectrum of one or more of the fluorescent tags, generates fluorescence with wavelengths within the emission spectrum of these fluorescent tags. This fluorescent l ight signal is captured in the sensing-wel l and directed to an optical detector through interference fi lters that ' fi lters out' most of the remaining laser l ight, thus allowing the measurement of only the desired fluorescent light.
[000408] In a preferred embodiment of this invention, a mix of two different capture molecules are immobil ized in each of the sensing wel ls. The mix contains, at a given ratio (e.g. 90: 10), a capture antibody specific to the target being detected and a second capture antibody which is specific to a control / normal izing moiety. While performing the test, a known concentration of the control / normalizing moiety is added to the sample, together with two detection antibodies, one specific to the target being detected and the other specific to the control / normalizing moiety. The two detection antibodies are each fluorescently labelled with a different fluorescent tag, each tag emitting at a different fluorescence wavelength. The binding to the surface of the sensing wells is monitored in given time-intervals (e.g. every 5 seconds) by shining laser-light from two different lasers, one after the other, into the excitation waveguides of the sensing chip. As a result, we get two different readings on the optical detector for every time interval. The first reading appears upon coupling laser # 1 into the excitation waveguides and it is a measure of the fluorescence emitted by the labelled detection antibodies specific to the target being detected. The second reading appears upon coupling laser #2 into the excitation waveguides and it is a measure of the fluorescence emitted by the labelled detection antibodies specific to the control / normalizing moiety. At each point in time, the first reading is normal ized to the second reading by dividing one by the other or by implementing any other suitable mathematical algorithm, leading to the elimination or reduction of aberrations caused by factors (such as temperature changes and reagents aging) which simi larly affect the target and the control moiety. In some embodiments, the time interval using for the first laser-light may be different than the interval used for the second laser-light. For example, one of the fluorophores may be more susceptible to photo-bleaching than the other fluorophore, so longer time intervals can be used with the photo-sensitive fluorophore in order to reduce photo- bleaching.
[000409] In a second preferred embodiment of this invention, a mix of two different capture molecules are immobi lized in the each of the sensing wel ls. The mix contains, at a given ratio (e.g. 90: 10), a capture antibody specific to the target being detected and a second capture antibody which is specific to a control / normal izing moiety. The test is performed in two steps. In the first step, a fluorescently label led detector antibody specific to the target is added to the sample at a known concentration and the mix is presented to the sensing chip. The binding to the surface of the sensing wells is monitored in given time- intervals (e.g. every 5 seconds) by shining laser-light from a single laser, into the excitation waveguides of the sensing chip. As a result, we get the time dependent fluorescent signal for the first step on the optical detector. In the second step, the control / normal izing moiety together with its specific fluorescently labelled detector antibody are added to buffer or other selected matrix (e.g. serum, plasma, oral fluid) at a known concentration and the mix is presented to the sensing chip. The binding to the surface of the sensi ng wel ls is monitored in given time-intervals (e.g. every 5 seconds) by shining laser- l ight from the same single laser, into the excitation waveguides of the sensing chip. As a result, we get the t ime dependent fl uorescent signal for the second step on the opt ical deteclor. The results of the first step are normal ized at the end of the second step by the results of the second step. The normalization can be done by dividing point-by-point or by dividing each point of the first step by the average or slope (over time) of the second step, or by dividing the signal slope (over lime) of the first step by the slope (over time) of the second step or any other mathematical algorithm that helps in the elimination of aberrations caused by factors (such as temperature changes and reagents aging) which similarly affect the target and the control moiety.
[000410] In a third preferred embodiment of the invention, a single capture molecule is immobilized in the each of the sensing wells. The capture molecule is specific to the target being detected. The test is performed in two steps. In the first step, a fluorescently labelled detector antibody specific to the target is added to the sample at a known concentration and the mix is presented to the sensing chip. The binding to the surface of the sensing wells is monitored in given time-intervals (e.g. every 5 seconds) by shining laser-light from a single laser, into the excitation waveguides of the sensing chip. As a result, we get the time dependent fluorescent signal for the first step on the optical detector. In the second step, the target moiety together with its specific fluorescently labelled detector antibody are added to buffer or other selected matrix (e.g. serum, plasma, oral fluid) at a known concentration and the mix is presented to the sensing chip. The binding to the surface of the sensing wells is monitored in given time-intervals (e.g. every 5 seconds) by shining laser-light from the same single laser, into the excitation waveguides of the sensing chip. As a result, we get the time dependent fluorescent signal for the second step on the optical detector. The results of the first reading are normal ized at the end of the second step by the results of the second reading. The normalization can be done by dividing point-by-point or by dividing each point of the first step by the slope (over time) of the second step, or any other mathematical algorithm that helps in the elimination of aberrations caused by factors (such as temperature changes and reagents aging).
[000411] In yet another preferred embodiment of this invention, a mix of three different capture molecules are immobil ized in the each of the sensing wells. The mix contains, at a given ratio (e.g. 80: 10: 10), the capture antibody specific to the target being detected and a two other capture antibodies which are each specific to one control / normalizing moiety. While performing the test, a known concentrations of the two control / normal izing moieties, preferably one at low concentration and the other at high concentration, are added to the sample, together with three detection antibodies, one specific to the target being detected and the two other specific to the two control / normalizing moieties. Using two or more normalization points al lows the generation of a cal ibration line or curve that reflects more accurately the effect that is being normalized for across the range measured, particularly when the effect, such as temperature or aging reagents, affects low concentrations differently than high concentrations. In some embodiments, the low concentration and the high concentration can be chosen to span the entire range to be measured. In some embodiments, the low concentration can be within 10, 20, 30, 40, or 50% of the low end of the range to be measured, and the high concentration can be within 10, 20, 30, 40, or 50% of the high end of the range to be measured. The three detection ant ibodies are each fl uorescently labelled with a different fl uorescent tag. each tag emitting at a di fferent fluorescence wavelength. The binding to the surface of the sensing wel ls is monitored in given time-intervals (e.g. every 5 seconds) by shining laser-light from three different lasers, one after the other, into the excitation waveguides of the sensing chip. As a result, we get three different readings on the optical detector for each time interval. The first reading appears upon coupling laser # 1 into the excitation waveguides and it is a measure of the fluorescence emitted by the labelled detection antibodies specific to the target being detected. The second reading appears upon coupling laser #2 into the excitation waveguides and it is a measure of the fluorescence emitted by the first labelled detection antibody specific to the first control / normalizing moiety. The third reading appears upon coupl ing laser #3 into the excitation waveguides and it is a measure of the fluorescence emitted by the second label led detection antibody specific to the second control / normalizing moiety. At each point in time, the signal from the two control / normal izing moieties are used for normalizing the first reading using a suitable mathematical algorithm.
Alternatively, at the end of test, the signal from the two control / normalizing moieties are used to generate a 'calibration curve / line' which is used to calculate the unknown concentration of the target.
[000412] Software based Auto-alignment of cartridge with optical head
[000413] In some embodiments, the substrate 804 of the sensing chip as shown in FIG. 8, for example, can be incorporated into a cartridge with a fluid port for introducing a sample, a flow path for directing the sample to the substrate 804, and a waste reservoir for collecting the sample after is passes over the substrate, as further described in U. S. Patent Publication No. 20160033412, which is herein incorporated by reference in its entirety. When the substrate 804/cartridge is inserted into the reader, it is aligned with the detector 806, which can be an optical laser head.
[000414] As shown in FIG. 7H, the substrate can be modified to include a loop back waveguide 713. A software and control system can be used to align the cartridge with the optical head.
[000415] When the cartridge is inserted, a sensor detects the insertion of the cartridge and triggers the light source to send a signal to the substrate and begin the alignment process. In some embodiments, the sensor can be a mechanical, electrical, or optical switch that is triggered when the cartridge is inserted into the reader.
[000416] The light travels through the loop back waveguide and a detector, such as a CMOS detector, at the other end of the loop back waveguide 71 3 receives the light.
[000417) If the cartridge is aligned properly or partially aligned, the detector receives the light transmitted through the loopback waveguide. The amount of light received determines the status of the al ignment.
[000418 | If the amount of l ight crosses a predetermined threshold for complete alignment, the alignment process stops and is complete and the system is ready for the actual testing of the sample.
[000419] When the system is not properly al igned, initially the system uses broad passes of the optical head to rapidly detect the location of the loopback channel . Using a closed loop feedback control system the auto-al ignment software controls the X & Y axis motion of the optical head and repeats this process til l the al ignment is done. I n case of partial light detection, a smart algorithm is developed to optimize the al ignment through X Y movement of the optical head ti l l complete al ignment happens.
[000420] Time outs can be put in place to detect error condit ions. [000421 ] A home position can be provided to bring the head to a fixed location in case the process has to be restarted.
[000422] With this automated system, no human intervention is needed to align the cartridge within the reader, making it a robust system.
[000423] FIG. 23 i llustrates a flowchart of an embodiment of the automated alignment procedure that can be implemented through a control system and/or software in the reader. In step 2300, a cartridge is inserted into a reader. As shown in step 2302, the reader system can ensure that the sample does not flow through the cartridge until after al ignment is successfully completed. This can be accomplished, for example, by not applying a vacuum or activating a pump until al ignment is successfully completed. After the cartridge is inserted into the reader, as shown in step 2304, light is sent to one end of the loop back waveguide of the substrate. A l ight detector at the other end of the loop back waveguide is used to measure the amount of light that is entering the loop back waveguide. As shown in step 2306, if the light measured by the loop back waveguide detector is greater than a predetermined level or at a maximum, the alignment is complete 2308. If the light measured by the detector is less than the predetermined level or less than a maximum detected level, then the optica! head that is providing the light to the substrate is moved in the X and/or the Y direction by a predetermined amount, which may be on the order of microns, as shown in step 23 10. The procedure then returns to step 2304, where light is again sent to the loop back waveguide and measured with the detector. In some embodiments, the optical head can be moved along a single axis, such as the X direction, until alignment is achieved along the X axis, and then the optical head can be moved along the second axis, such as the Y axis, until alignment is achieved along the Y axis. In some embodiments, the increase or decrease in light measured by the detector as the optical head is moved in a particular direction can be used to determine whether the optical head is being moved in the correct direction for achieving alignment. An increase in measured light indicates that the movement of the optical head is proceeding in the correct direction, whi le a decrease in measured light indicates that the movement of the optical head is proceeding in the wrong direction. In some embodiments, the optical head can be alternately moved in the X and Y directions during alignment.
[000424] Precision in printing antibodies on the detection chip
[000425] Very small volumes of antibodies in solution (-200 Pico liters) can be dispensed as droplets on the optical sensing sites of the substrate with high precision and high accuracy as described below. Dispensing of multiple layers of different samples, antibodies, ligands, oligonucleotides, or reagents on a plural ity of locations on a substrate is a significant chal lenge as it requires greater level of precision and flexi bility. Although the printing method is described with antibodies, other molecules, reagents, proteins, ligands, oligonucleotides, etc. can be also printed.
|000426] A use of specialized printer with hardware and software that supports a non-contact method of printing antibodies and a method to dry, detect and test the micro-structures formed by loading the antibody droplets on the specific spots, such as the sensing sites, on the bio-sensor chip, also called more generically as a substrate. |000427] Special software is used to do on-line measurement of droplet volumes. A microscope with CCD camera is used to take images of the dispensed antibodies on the detection spots. A special image analysis software is used for spot detection, analysis of microstructures and post spotting Quality Control, making sure that the size and shapes and location of the spots are per specification and that the adjacent spots don't overlap with each other, which may create interference in the detection results.
[000428] FIG. 24 i llustrates a flowchart of the antibody printing procedure. In step 2400, droplets of antibodies are disposed onto a substrate using a plurality of droppers arranged in array. In some embodiments, the array is l inear. In other embodiments, the array can be two dimensional. As noted above, the droppers can dispose the droplets in a non-contact manner, meaning that the droplets are disposed above the surface of the substrate and then fall onto the substrate. The droppers never contact the substrate. In step 2402, a high resolution and high precision image of the droplets on the substrate can be automatical ly taken. In step 2404, image processing software can be used to analyze the image to determine the size, volume, shape, and location of the droplets which can be used in step 2406 to determine whether all the antibody droplets are placed correctly on the substrate. If the droplets are placed correctly, then the substrate can be kept for use and further processing in step 2408. If the droplets are not placed correctly, then the substrate can be thrown away in step 2410.
[000429] FIGS. 25A-25D illustrate a linear array of droppers 2500 that are dispensing droplets 2502 onto a substrate 2504. The linear array of droppers are part of a specialized printer 2506 and imager.
EXAMPLES
Example 1
Establishing the Limit of Detection
[000430] One feature of the sensors of the current invention is the ability to detect low numbers of biologically active analytc molecules. The following experiments demonstrate this capability. The limit of detection (LOD) of the sensors was determined by measuring fluorescently labeled ovalbumin in solution. The dye used was Alexa Fluor 660; it was excited at 658 nm and emission was measured at 690 nm. All measurements were made at room temperature (25° C). LOD was assayed using two different methods: 1 ) determination of the lowest concentration of labeled oval bumin detectable above buffer background with better than 95% confidence (P<0.05, Student's t-test); and 2) determination of the analytical sensitivity from a standard curve of sensor response vs. concentration (MDL=2*SD/slope, where SD is the standard deviation of the buffer background measurement and slope is the initial slope of the standard curve). These methods are comparable in that both determine the 95% confidence level for LOD. Values for LOD from three different step isotherms are shown in Table 1 . TABLE 1
MDL MDL
Isotherm Chip Format Method 1 Method 2
Experiment 1 100 x 1 00 μιη sensors 3.0 pM 4.1 pM
Experiment 2 52.5 x 4,500 μιπ sensors 0.1 pM 0.33 pM
Experiment 3 52.5 x 4.500 μηι sensors 0.1 pM 0.13 pM
[000431] The number of molecules detected per sample was calculated as follows. The sample volume used in all experiments was 50 microliters. Thus, at a concentration of 0.1 pM for example, there were 3 x l 06 molecules per sample. Since all molecules were evenly distributed across the entire volume, the number of molecules available per sensor was ~1000. The thickness of the fluid containing this number of molecules was ~1 .7 mm. Using the diffusion parameters, it can be estimated that 0.1 % to 1 % of these molecules were able to bind to the surface of each sensor, or 1 to 10 molecules.
Example 2
Detection of Protein-Protein Interactions
[000432] The present invention is useful for measuring protein-protein interactions. In this example, the waveguide-based optical detection technology was used to measure the binding of ovalbumin to anti-ovalbumin antibodies. Before the experiment was conducted, chips with 52.5x4,500 pm sensing elements were dip-coated with anti-ovalbumin antibodies. This procedure involved several steps. First, the chips were cleaned. A chip fabricator coated the active surface of the chips with a thin polymer layer to protect it during wafer dicing. The polymer layer was removed by immersing chips in acetone for 5 minutes, followed by isopropanol for 5 minutes and a second isopropanol rinse for an additional minute. Chips were then washed in deionized water three times and dried in a vacuum desiccator. Next, the surface was activated. To immobilize capture molecules to the waveguides, a thin layer (~10 nm) of silicon dioxide was created on the chips by Piranha treatment. Chips were immersed in a Piranha solution containing 9% (v/v) H202 and 66.5% (v/v) H2S04 for 45 minutes with agitation. This step removed organic contaminants and produced a thin ( 1 -2 nanometer) layer of reactive silanol groups that were used for coupling antibodies. Chips were then rinsed four times in deionized water, followed by a final rinse in doubled distil led water.
[000433] Anti bodies specific to ovalbumin were immobi lized to the chips using an avidin/biotin chemistry that forms self-assembl ing monolayers. (Herron, J. N., H.- . Wang, V. Janatova, J . D.
Durtschi, . D. Caldwell. D. A . Christensen, I .-N. Chang and S.-C. Huang (2003) Orientation and Activity of Immobi l ized Antibodies. In: Biopolymers at Interfaces, 2nd Edition (M. Malmsten, ed.), Surfactant Science Series. Vol. 1 1 0, Marcel Dekker, new York, pp. 1 15- 163.) Avidin can adsorb to surfaces containing silanol groups due to electrostatic interactions. Antibody immobil ization proceeded in two steps: adsorption of avidin to the Piranha treated chips ( 1 00 iiM avidin in phosphate buffered saline, pH 7.4. adsorption time of 1 hr. fol lowed by five PBS rinses) followed by binding of biotinylated anti- ovalbumin antibody (e.g. US Biological, polyclonal) to the adsorbed avidin ( 100 nM biotinylated anti- ovalbumin antibody in PBS). Chips were then rinsed three times in PBS, five more times in deionized water, then postcoated with a xeroprotectant (0.1 mg/mL trehalose in deionized water) and dried by nitrogen stream, followed by vacuum desiccation.
|000434] It was estimated that the capture antibody density at the end of the process was 1 p per sc]uare centimeter.
[000435] To assay the interaction of ovalbumin with the anti-ovalbumin antibodies, fluorescently labeled ovalbumin in a solution containing phosphate buffered saline, pH 7.4, and 0.1 mg/mL bovine serum albumin, was added to a sample well on the surface of a chip as depicted in FIG. 7D. An ovalbumin concentration range of four orders of magnitude (from 0.1 pM to 1 ,000 pM) was used. The kinetics of the binding reaction were then monitored over a period of 10 minutes at room temperature, and fluorescence intensity was measured at 690 nm.
[000436] FIG. 19 shows the quantitative sensor response for the binding reaction. Fluorescence intensity at each ovalbumin concentration used is indicated with plot symbols and error bars and is plotted on a graph with log based abscissa and ordinate axes. The solid line in FIG. 19 is a curve fit. A two- constant model fit the binding data, representing binding behavior that is consistent with results from experiments using ultrasensitive (femtomolar) sensors in conjunction with l -pm thick excitation waveguides (Plowman, T. E., W. M. Reichert, C. R. Peters, H. . Wang, D. A. Christensen and J. N. Herron ( 1996), "Femtomolar Sensitivity using a Channel-etched Thin Film Waveguide
Fluoroimmunosensoiy' Biosensors & Bioelectronics 1 1 , 149-160). A small percentage of ovalbumin molecules (0.22%) simultaneously bound two different immobilized antibody molecules, while the overwhelming majority (>99%) of ovalbumin molecules bound to a single immobilized antibody. The small percentage of bivalent binding gives rise to the higher affinity binding constant (K,), as well as the "foot'" observed in the subpicomolar portion of the binding curve. Only a small percentage of high affinity binding was observed owing to the exquisite sensitivity and excellent precision of the sensor in the subpicomolar range. Precision was generally good across the 4-log concentration range of the assay.
Example 3
Detection of Polymerization of Nucleic Acids
[000437] The waveguide-based optical detection system can be used to quantify the polymerization of DNA in a primer extension assay. In a primer extension reaction, an mRNA target hybridizes to a DNA primer, and reverse transcriptase uses the mRNA target as a template to add deoxynucleotides (dATP, dTTP. dGTP, dCTP) to the 3' end of the DNA primer.
[000438] High sensitivity using a waveguide-based optical detection system requires that the hybridization in the primer extension reaction occur at concentrations in the low femtomolar range. Thus, it was first establ ished that hybridization occurs at low concentrations on the surface of the chip. For this experiment, a capture ol igonucleotide primer was immobilized to the chip, and a complementary synthetic DNA labeled at the 5 ' end with Cy5.5 was incubated with the primer. For experimental rigor. the sequences of both the captured oligonucleotide and a complementary synthetic DNA were verified by mass spectrometry.
[000439] Before the experiment was conducted, the capture oligonucleotide was immobilized on the surface of the chip by a dip-coating process. First, the chips were cleaned. A chip fabricator coated the active surface of the chips with a thin polymer layer to protect it during wafer dicing. The polymer layer was removed by immersing chips in acetone for 5 minutes, followed by isopropanoi for 5 minutes and a second isopropanoi rinse for an additional minute. Chips were then washed in deionized water three times and dried in a vacuum desiccator.
[000440] Next the surface was activated. To immobilize capture molecules to the waveguides, a thin layer (~10 nm) of silicon dioxide was deposited on the chips by either Piranha treatment or oxygen plasma treatment. Piranha treated chips were immersed in a Piranha solution containing 9% (v/v) H202 and 66.5% (v/v) H2S04 for 45 minutes with agitation. This step produced a thin (1 -2 nanometer) layer of reactive silanol groups. Chips were then rinsed four times in deionized water, followed by a final rinse in doubled distilled water. Oxygen plasma treated chips were exposed to oxygen plasma ( 100W, 0.15 Torr) for 5 minutes using a plasma oven.
[000441] To attach oligonucleotides, activated chips (either Piranha or plasma treated) were derivatized with (3-Glycidyloxypropyl) trimethoxysilane (GPS) to form a reactive monolayer of epoxide groups on the surface of the chip. Chips were immersed in 0.1 % (v/v) GPS in anhydrous toluene for 30 minutes at 40° C, followed by washing three times in 99.9% toluene. Chips were then dried in nitrogen and cured at 1 10° C. for 20 minutes. GPS-coated chips were immersed for 6 hours at 40° C. in a 5 micromolar solution of capture oligonucleotide dissolved in 0.1 M carbonate-bicarbonate buffer (pH 9) with 1 mM EDTA . The capture oligonucleotides were synthesized with a flexible spacer at the 5' end that terminated in a primary amino group, which reacts readily to the epoxy groups on the chip to form a stable covalent linkage. Chips were washed after the coupling reaction in 0.2% SDS, followed by three rinses in deionized water. Chips were then incubated in deionized water at 40° C. for 30 minutes and then dried in a nitrogen stream followed by vacuum desiccation overnight.
[000442] It was estimated that the density of capture oligonucleotides at the end of the process was 1 0 p per square centimeter.
[000443] Hybridization reactions were performed for 10 minutes at room temperature over a synthetic D1MA analyte concentration range of 10 femtomolar ( 10 f ) to 10 picomolar ( 10 pM). Analyte sol utions were prepared i n 3 X sodium chloride-sodium citrate buffer (0.45 M NaCl, 45 mM sodium citrate, pH 7) with 0.01 % sodium dodecyl sulfate and 1 mM EDTA. Limit of detection (LOD) was assessed using analysis of variance (ANOVA) with Dunnett's post-hoc test in which hybridization data from each concentration were compared to that of the negative control (buffer). Results are shown in Table 2 for two different experiments (Experiment 1 and Experiment 2): TABLE 2
Dunnett's Multiple Comparison
Iql
Comparison Mean Difference (Dunnett's Statistic) Probability
Experiment 1
Buffer vs 10 fM -2.58 1.53 0.4881
Buffer vs 30 fM -6.66 3.94 0.0008
Buffer vs 1 00 fM -9.1 5 5.41 < 0001
Buffer vs 300 fM -21 .3 12.6 <.0001
Buffer vs 1 pM -29.1 17.2 < 0001
Buffer vs 3 pM -37.7 21 .0 < 0001
Buffer vs Ι Ο ρΜ -147 86.7 <.0001
Experiment 2
Buffer vs 1 0 fM - 19.5 2.30 0.1 148
Buffer vs 30 fM -40.5 4.78 0001
Buffer vs 100 fM -53.1 6.27 < 0001
Buffer vs 300 fM - 1 1 5 13.6 0001
Buffer vs 1 pM -93.9 1 1 .1 < 0001
Buffer vs 3 pM - 126 14.9 < 0001
Buffer vs 1 0 pM -305 36.0 < 0001
|000444] Mean Difference values were negative because the sensor response for each
concentration was subtracted from that of the negative control (Buffer). Probabil ity values are for the null hypothesis that the sensor response observed for a given concentration is not statistically different from that observed for the negative control. Using P<0.05 as the criterion for rejecting the null hypothesis, concentrations of 30 fM and above were statistically different from buffer in both experiments (i.e., MDL=30 fM). Comparison of the P values for the 10 fM concentrations in Experiments 1 & 2 (P=0.4881 for Experiment 1 and P=0.1 1 48 for Experiment 2) show that sensitivity was better in the second experiment, approaching 10 fM.
[000445] Recycling chips by stripping off their layer of capture molecule with the aforementioned hydrogen peroxide/sulfuric acid cleaning solution and then immobil izing a new layer of capture molecules once resulted in diminished, but measurable hybridization levels with reduced sensitivity (in the low picomolar range; data not shown).
[000446] A primer extension experiment was conducted using an RNA standard (kanamycin control RNA) purchased from Promega, Inc. The capture oligonucleotide primer was complementary to a 24 base sequence near the 3' end of the RNA standard. This sequence is located 971 bases downstream from the 5' end of the RNA standard and contains 224 guanines. The cytosine deoxynucleotides were labeled with a fluorescent dye (Cy5.5). Roche's Transcriptor Reverse Transcriptase was used to extend the primer. The primer extension reaction was carried out at 48° C, which yielded reduced non-specific hybridization. Results from primer extension of 1 pM kanamycin RNA are shown in FIG. 20. The extension rate for the control is slightly negative (-9.7 mV/min), which is probably due to instrument noise. The extension rate of 1 pM kanamycin RNA is 128 mV/min, significantly higher than that of the control. Moreover, the signal increases linearly over the 1 0.5-minute reaction period with no sign of saturation. The initial signal level of the 1 pM RNA reaction was somewhat lower than that of the control run, and potentially due to a temperature difference ( 1 -2° C.) between the two runs.
Example 4
Quantitative Real-Time PCR Detection of Transcripts
[000447] Quantitative real-time PCR detection of transcripts, for example, bcr/abl fusion transcripts, can be achieved using the detection system of the invention, using a PCR assay protocol modified from that described by Kreuzer et al. (supra). The system includes a substrate chip with excitation waveguides, collection waveguides and intersection regions. The intersection regions where the excitation waveguides and collection waveguides cross include optical sensing sites with sensing wells for conducting quantitative real-time PCR. The detection system further includes a scanning light source which is coupled to and in optical communication with the excitation waveguides of the substrate at some point along its scanning path. The system further includes an optical detector.
[000448] Samples suspected of containing RNA transcripts of interest (e.g., blood from a subject) are treated to obtain a source of total RNA which is subsequently reverse transcribed using reverse transcriptase into cDNA using techniques well known in the art. cDNA samples are delivered to sensing sites on the substrate of the system. As desired, suitable controls and different dilutions of a particular cDNA sample can be delivered to different sensing wells.
[000449] Reagents for real-time reverse transcriptase PCR are provided at the sensing sites. PCR reactions are conducted using primers/probes specific for one or more bcr/abl breakpoint cluster region. Such primers/probes are well known in the art (e.g., see Kreuzer et al. supra) and can be labeled with 6- carboxy-fluorescein phosphoramidite at the 5' end, and as a quencher, 5-carboxy-tetramethyl-rhodamine can be incorporated further along the primer/probe sequence. As the primer/probe is hydrolyzed through the 5'-nucelase activity of Taq DNA polymerase, unquenched fluorescence from the fluorescein (reporter) dye can be induced. Phosphate groups are attached to primers/probes 3' end to prevent probe extension. A 1 0-μΙ PCR reaction mix contains 1 μΐ of 10x PCR buffer, 4.5 mM gC12, 0.8 ni dNTP, 0.5 μ each primer. I μΜ probe, 0.2 units of a temperature-release Taq DNA polymerase (Platinum® Pfx DNA Polymerase; Invitrogen, Corp.), and 20 ng of sample cDNA. PCR ampl ification is started with a 5-min denaturation step at 94° C, followed by 45 cycles of denaturation at 94° C. for 30 s and
anneal ing/extension at 65° C. for 60 s.
[000450] While PCR ampl ification proceeds excitation light at a wavelength of about 658 nm is generated by the scanning light source and is directed lo each sensing well by the excitation waveguides. If the cDNA samples include a transcript for bcr/abl fusion transcripts, annealing of primers/probes to the cDNA should occur. Subsequent hydrolysis of the probe by polymerase and unquenching of the fluorescein reporter results in fluorescence in the sensing well. As the number of cycles increases, the amount of unquenched fluorescein increases in relation to the amount of bcr/abl fusion transcript cDNA in the reaction.
[000451 ] By way of the collection waveguides fluorescence in the sensing wells is detectable by the optical detector of the system. Thus, detection of bcr/abl fusion transcripts can be measured in realtime and based on appropriate controls and analysis the amount of bcr/abl fusion transcripts in a sample can be quantified.
Example 5
Detection of HIV Status of Multiple Subjects
[000452] Fluorescent immunoassay-based detection of HI V+ status of multiple subjects can be achieved using a detection system such as the example illustrated in FIG. 7A. The system includes a substrate chip with excitation waveguides, collection waveguides and intersection regions. The intersection regions where the excitation waveguides and collection waveguides cross include optical sensing sites with sensing wells for conducting fluorescent immunoassays. The detection system further includes a scanning light source which is coupled to and in optical communication with the substrate at some point along its scanning path and the excitation waveguides. The system further includes an optical detector.
[000453] A partially purified antigen, for example, inactivated HIV protein p29 antigen, is pre- coated onto the sensing wells of the optical sensing sites. Next, a number of subject serums which may contain antibodies to HIV p29 are delivered to separate sensing sites. It is also envisioned that samples from multiple patients are pooled and that each pool is delivered to separate sensing sites. If a subject is H I V+, then their serum may contain antibodies to HIV protein p29, and those antibodies will bind to the H I V p29 antigens on the sensing sites. After a washing step, anti-human immunoglobul in coupled to a fluorescent dye (fluorescein) is added to the sensing sites. This secondary antibody binds to human antibodies in the sensing sites (i.e., the anti-p29 antibodies). Next an excitation l ight at a wavelength of about 658 ran is generated by the scanning light source and is directed to each sensing well by the excitation waveguides. If the secondary antibody is present the coupled fluorescein will fluoresce in the presence of the excitation light in the well.
[000454] By way of the collection waveguides fluorescence in the sensing wells is detectable by the optical detector of the system. Signal received by the optical detectors can be interpreted to determine i f a given subject, or a sample pooled from multiple subjects, has antibodies to H IV p29. Suitable controls can be used to validate the results of the assay. Thus, the presence or absence of HIV p29 can be measured in a sample and H 1V+ or HIV(-) status of multiple subjects or pools can be determined. Example 6
Two-Site Sandwich Immunoassays
[000455] A two-site sandwich immunoassay can be employed in assays using the detection system of the invention (e.g., the systems as illustrated in FIG. 7A-7E). Antibodies fulfi ll two different roles in such assays, in which an immobilized antibody captures the analyte, while a soluble, fluorescently labeled antibody detects or "traces" analyte binding. To prevent competitive binding, capture and tracer antibodies must bind to different sites on the analyte molecule. For large analytes with repetitive epitopes (e.g., viruses and bacteria), a single antibody (specific for the repetitive epitope) can usually be employed in both capture and tracer roles. Smaller analytes (e.g. proteins and polysaccharides) expressing multiple, unique epitopes require two different antibodies, each specific for a unique epitope.
[000456] Three two-site sandwich immunoassay tests are envisioned: 1 ) serial testing of optical sensing sites; 2) low complexity parallel testing of optical sensing sites; and 3) sensitivity testing. In the first of these, a small volume (1 -5 μΙ_.) of sample (containing analyte and tracer antibody) is spotted directly at a optical sensing site containing capture antibody using a microliter pipette. Binding kinetics at the site are monitored over a 5-min period at room temperature. Translation of optical detection to excitation and collection waveguides in connection with a different optical sensing site is effected and the assay is repeated at the new site. It is envisioned that at least 10 optical sensing sites can be tested using this serial procedure. Such tests can demonstrate sensitivity and intra-assay precision of the system.
[000457] In the second form of testing, using a substrate of the system that includes a 10x 10 array of excitation waveguides and collection waveguides (see e.g., FIG. 7A), a single excitation waveguide of the substrate is excited, while output signals are monitored from all 10 collection waveguides using a linear detector array. Equal volumes (e.g., 50 μΕ) of sample containing analyte and tracer antibody (e.g., 10 n , final concentration) can be pre-mixed and then injected into a sample well of the optical sensing site that contains capture antibody. Binding kinetics are monitored simultaneously in 10 optical sensing sites over a 5-min period at room temperature. This form of testing can demonstrate the parallel assay capabilities of the system, as well as providing more detailed information about intra-assay precision.
[000458] In the third form of testing, the sensitivity, precision and linearity of the device can be demonstrated by constructing a standard curve of average reaction rate versus analyte concentration. Device configuration is the same as described above for the second form of testing (i.e., 10 simultaneous assays). Analyte concentration is varied over at least a 1 00-fold range, e.g. 1 0 pM to 1 nM, though the exact range can be adjusted depending on the clinical concentration range of the analyte being examined. A separate substrate chip is used for each concentration to be tested. Six to eight concentrations are examined. Resulting standard curves are typically linear at low concentration, but saturate at higher concentrations.
[000459] The Herron lab has developed immunoassays for many different analytes including human cardiac troponin 1 (cTnl), chorionic gonadotrophin (hCG), creatine phosphokinase isoform M B (CKM B), myoglobin, ovalbumin (used by the mi l itary as a "simulant" for toxins such as ricin and SEB). ricin. Staphylococcal enterotoxin B (SEB). (See Herron, J. N. et al . (2003). Orientation and Activity of I mmobilized Antibodies. In: Biopoiymers at Interfaces, 2nd Edition (M. Malmsten, ed.), Surfactant Science Series, Vol. 1 10, Marcel Dekker, New York, pp. 1 1 5-163; and Herron, J. N. et al. (2005). Planar Waveguide Biosensors for Point-Of-Care Clinical and Molecular Diagnostics. In: Fluorescence Sensors and Biosensors (R. B. Thompson, Ed.), CRC Press Taylor & Francis Group, Boca Raton, Fla. pp. 283- 332).
[000460] The ovalbumin assay of Herron can be used in the first and second immunoassays described above. Advantageously, reagents for this assay are relatively inexpensive and no special handling is required. Detection requirements for cTnl and SEB are the most stringent, and thus immunoassays specific for these analytes are useful for the sensitivity testing immunoassays. However, since the CDC, NIH, and USDA all list SEB as a select agent requiring special handling it may be preferable to use cTnl in sensitivity testing. cTnl can be paired with two other cardiac markers (C MB and myoglobin) for simultaneous immunoassay sensitivity testing.
[000461] Detection of Clostridium difficile toxin A was used in the second and third immunoassays described above to demonstrate the rapidity and sensitivity of the detection system of the current invention. FIG. 21 depicts a real-time detection ofClostridiwn difficile toxin A in bovine serum on a 1 0- channel chip. In the first 5 minutes (300 seconds) no analyte is present and thus the signal is flat. After 5- minutes analyte is added to reach a concentration of 10 pM. As the analyte binds to the capture antibody immobilized to the chip surface, and a second labeled antibody binds to the complex in a sandwich assay format, the optical signal in all channels increases. The slope of the increasing signal is proportional to the analyte concentration.
[000462] FIG. 22 shows a standard curve for Clostridium difficile toxin A as measured on a series of ten 10-channel chips. The optical signal slope increases as a function of the increasing concentration. Ten different concentrations were measured, starting from 0.1 pM and up to 1 μΜ. As can be seen, the slope concentration relation is linear between 0.3 pM and 1 nM. The limit of detection (LOD) is approximately 0. 1 pM. These results demonstrate significant improvements in rapidity and sensitivity as compared to commercially available tests for Clostridium difficile toxin A, which have assay times ranging from 1 5 to 75 minutes and LODs ranging from 2.7 to 133 pM.
[000463] While preferred embodiments of the present invention have been shown and described herein, it wi ll be obvious to those skilled in the art that such embodiments are provided by way of example only. Numerous variations, changes, and substitutions will now occur to those skilled in the art without departing from the invention. It should be understood that various alternatives to the
embodiments of the invention described herein may be employed in practicing the invention. It is intended that the following claims define the scope of the invention and that methods and structures within the scope of these claims and their equivalents be covered thereby.

Claims

CLAIMS What is claimed is:
1 . A detection system for detecting a biologically active analyte molecule comprising:
a substrate comprising one or more excitation waveguides, a plurality of collection waveguides, the one or more excitation waveguides and the plurality of col lection waveguides crossing to form an array of intersection regions where an excitation waveguide and a collection waveguide cross and provide optical communication with the intersection region at each crossing, and a plurality of optical sensing sites each in optical communication with an intersection region;
a scanning light source, wherein the scanning light source is at some point along its scanning path in optical communication with at least one of the one or more excitation waveguides;
a detector that is in optical communication with one or more of the collection waveguides; and an actuator for spatially translating a light beam emitted from the scanning light source relative to the substrate such that the light beam is coupled to and in optical communication with at least one of the one or more excitation waveguides of the substrate at some point along its scanning path.
2. The detection system of claim 1 , wherein the one or more excitation waveguides is a single excitation waveguide.
3. The detection system of claim 1 , wherein both the one or more excitation waveguides and the plurality of collection waveguides exit the substrate at a first edge of the substrate.
4. The detection system of claim 3,
wherein the scanning light source is at some point along its scanning path in optical
communication with one or more of the excitation waveguides at the first edge of the substrate; and
wherein the detector is in optical communication with one or more of the collection waveguides at the first edge of the substrate.
5. The detection system of claim 1 , wherein the one or more excitation waveguides are curved about 90 degrees.
6. A detection method comprising:
delivering a sample suspected of containing a biological ly active analyte molecule to be detected to an optical sensing site on a substrate of a detection system, the substrate comprising one or more excitation waveguides and a plurality of collection waveguides; spatially translating a scanning light source to a point at which the light source is in optical communication with at least one of the one or more excitation waveguides, wherein at least one of the one or more excitation waveguides isjn optical communication with the optical sensing site, thereby generating a first l ight wave within said at least one of the one or more excitation waveguides, wherein the first light wave is transducable by a sensor associated with the optical sensing site to a second light wave carried in one or more of the plurality of collection waveguides in optical communication with the optical sensing site and crossing the one or more excitation waveguides; and
detecting a measurable change in the second light wave using a detector in optical
communication with one or more of the plurality collection waveguides, wherein a measurable change in the second light wave occurs when the sensor interacts with the biologically active analyte molecule.
7. The method of claim 6, wherein the scanning light source further comprises a detector, and wherein at the point at which the light source is coupled to and in optical communication with one or more waveguides in optical communication with the optical sensing site, the detector is also coupled to and in optical communication with said one or more waveguides.
8. The method of claim 6, wherein the biologically active analyte is selected from the group consisting of a nucleic acid, a protein, an antigen, an antibody, a microorganism, a gas, a chemical agent and a pollutant.
9. The method of claim 6, wherein the sensor is adapted to support an immunoassay and wherein the sensor interacting with the biological ly active analyte comprises an outcome of an immunoassay.
10. The method of claim 6, wherein the one or more excitation waveguides is a single excitation waveguide.
1 1 . The method of claim 6, wherein both the one or more excitation waveguides and the plurality of collection waveguides exit the substrate at a first edge of the substrate.
12. The method of claim 1 1 ,
wherein the scanning l ight source is at some point along its scanning path in optical
communicat ion with one or more of the excitation waveguides at the first edge of the substrate; and
wherein the detector is in optical communication with one or more of the col lection waveguides at the first edge of the substrate.
13. The method of claim 6, wherein the one or more excitation waveguides are curved about 90 degrees.
14. A method of measuring a concentration of an analyte using a waveguide chip, the method comprising:
adding a known amount of one or more normal izing moieties to a sample;
adding a first detection molecule labelled with a first fluorescent tag to the sample, the first detection molecule specific to the analyte;
adding one or more secondary detection molecules, each secondary detection molecule labelled with a distinct secondary fluorescent tag, to the sample, the one or more secondary detection molecules specific to the one or more normalizing moieties, wherein the first fluorescent tag and the one or more secondary fluorescent tags emit at different fluorescence wavelengths; introducing the sample to an array of sensing sites on the waveguide chip, each sensing site of the array having a first capture molecule specific to the analyte and one or more secondary capture molecules specific to the one or more normalizing moieties, wherein the first capture molecule and the one or more secondary capture molecules are both immobilized in each sensing site of the array at a predetermined ratio;
introducing a first l ight pulse into one or more excitation waveguides on the waveguide chip at a first predetermined interval, the one or more excitation waveguides in optical communication with the array of sensing sites, the first l ight pulse configured to excite the first fluorescent tag;
introducing one or more secondary light pulses into the one or more excitation waveguides on the waveguide chip at one or more secondary predetermined intervals, wherein the first light pulse and the one or more secondary light pulses have different wavelengths, the one or more secondary light pulses configured to excite the one or more secondary fluorescent tags; measuring the fluorescence emitted by the first detection molecule and the one or more secondary detection molecules through a plurality of col lection waveguides in optical communication with the array of sensing sites; and
normalizing the measured fluorescence emitted by the first detection molecule using the
measured fluorescence emitted by the one or more secondary detection molecules.
1 5. The method of claim 14, wherein the step of normal izing the measured fluorescence emitted by the first detection molecule comprises dividing the measured fluorescence emitted by the first detection molecule by the measured fluorescence emitted by the one or more secondary detection molecules.
16. The method of claim 14, wherein the fu st detection molecule, the one or more secondary detection molecules, the first capture molecule, and the one or more secondary capture molecules are antibodies.
17. The method of claim 14, wherein the step of normalizing the measured fluorescence emitted by the first detection molecule further comprises generating one or more calibration curves from the measured fluorescence emitted by the one or more secondary detection molecules.
18. The method of claim 14, wherein the one or more normalizing moieties comprises at least a first normalizing moiety and a second normalizing moiety, wherein the amount of the first normal izing moiety added to the sample is greater than the amount of the second normalizing moiety added to the sample.
19. The method of claim 14, wherein the step of normalizing the measured fluorescence emitted by the first detection molecule comprises dividing the measured fluorescence emitted by the first detection molecule by a slope of the measured fluorescence emitted by the one or more secondary detection molecules.
20. The method of claim 14, wherein the step of normalizing the measured fluorescence emitted by the first detection molecule comprises dividing a slope of the measured fluorescence emitted by the first detection molecule by a slope of the measured fluorescence emitted by the one or more secondary detection molecules.
21 . A method of measuring a concentration of an analyte using a waveguide chip, the method comprising:
adding a known amount of a first detection molecule labelled with a first fluorescent tag to the sample, the first detection molecule specific to the analyte;
introducing the sample to an array of sensing sites on the waveguide chip, each sensing site of the array having a first capture molecule specific to the analyte and a second capture molecule specific to the normalizing moiety, wherein the first capture molecule and the second capture molecule are both immobilized in each sensing site of the array at a predetermined ratio; introducing a first light pulse into one or more excitation waveguides on the waveguide chip at a first predetermined interval, the one or more excitation waveguides in optical communication with the array of sensing sites;
measuring the fluorescence emitted by the first detection molecule through a plural ity of
col lection waveguides in optical communication with the array of sensing sites; introducing a known amount of a normal izing moiety with a known amount of a second detection molecule label led with a second fl uorescent tag to the array of sensing sites, the second detection molecule specific to the normal izing moiety;
introducing a second l ight pulse into me one or more excitation waveguides on the waveguide chip at a second predetermined interval, the one or more excitation waveguides in optical communication with the array of sensing sites; measuring the fluorescence emitted by the second detection molecule through the plurality of collection waveguides; and
normalizing the measured fluorescence emitted by the first detection molecule using the
measured fl uorescence emitted by the second detection molecule.
22. The method of claim 21 , wherein the step of normalizing the measured fluorescence emitted by the first detection molecule comprises dividing the measured fluorescence emitted by the first detection molecule by the measured fluorescence emitted by the second detection molecule.
23. The method of claim 21 , wherein the step of normalizing the measured fluorescence emitted by the first detection molecule comprises dividing the measured fluorescence emitted by the first detection molecule by an average of the measured fluorescence emitted by the second detection molecule.
24. The method of claim 21 , wherein the first predetermined interval is the same as the second predetermined interval.
25. The method of claim 21 , wherein the first predetermined interval is different than the second predetermined interval.
26. The method of claim 21 , wherein the first light pulse and the second light pulse have the same wavelength.
27. A method of measuring a concentration of an analyte using a waveguide chip, the method comprising:
adding a known amount of a first detection molecule labelled with a first fluorescent tag to the sample, the first detection molecule specific to the analyte;
introducing the sample to an array of sensing sites on the waveguide chip, each sensing site of the array having an immobilized capture molecule specific to the analyte;
introducing a first l ight pulse into one or more excitation waveguides on the waveguide chip at a first predetermined interval, the one or more excitation waveguides in optical communication with the array of sensing sites;
measuring the fluorescence emitted by the first detection molecule through a plurality of
collection waveguides in optical communication with the array of sensing sites; introducing a known amount of the analyte with a known amount of a second detection molecule label led with a second fl uorescent tag to the array of sensing sites, the second detection molecule specific to the analyte; introducing a second light pulse into the one or more excitation waveguides on the waveguide chip at a second predetermined interval, the one or more excitation waveguides in optical communication with the array of sensing sites;
measuring the fluorescence emitted by the second detection molecule through the plurality of collection waveguides; and
normalizing the measured fluorescence emitted by the first detection molecule using the
measured fluorescence emitted by the second detection molecule.
28. The method of claim 27, wherein the step of normalizing the measured fluorescence emitted by the first detection molecule comprises dividing the measured fluorescence emitted by the first detection molecule by the measured fluorescence emitted by the second detection molecule.
29. The method of claims 21 or 27, wherein the step of normalizing the measured fluorescence emitted by the first detection molecule comprises dividing the measured fluorescence emitted by the first detection molecule by a slope of the measured fluorescence emitted by the second detection molecule.
30. The method of claims 21 or 27, wherein the step of normalizing the measured fluorescence emitted by the first detection molecule comprises dividing a slope of the measured fluorescence emitted by the first detection molecule by a slope of the measured fluorescence emitted by the second detection molecule.
3 1 . The method of claim 27, wherein the first light pulse and the second l ight pulse have the same wavelength.
PCT/US2016/055167 2015-10-01 2016-10-03 Waveguide-based detection system with scanning light source WO2017059425A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201562235977P 2015-10-01 2015-10-01
US62/235,977 2015-10-01
US15/245,062 US20170082617A1 (en) 2006-03-10 2016-08-23 Waveguide-based detection system with scanning light source
US15/245,062 2016-08-23

Publications (1)

Publication Number Publication Date
WO2017059425A1 true WO2017059425A1 (en) 2017-04-06

Family

ID=58427987

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2016/055167 WO2017059425A1 (en) 2015-10-01 2016-10-03 Waveguide-based detection system with scanning light source

Country Status (1)

Country Link
WO (1) WO2017059425A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10660619B2 (en) 2017-07-19 2020-05-26 Evanostics Llc Cartridges for oral fluid analysis and methods of use
US11262367B2 (en) 2017-12-15 2022-03-01 Evanostics Llc Optical reader for analyte testing
US11467086B2 (en) * 2019-08-30 2022-10-11 Canon Medical Systems Corporation Sample inspection apparatus

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5544268A (en) * 1994-09-09 1996-08-06 Deacon Research Display panel with electrically-controlled waveguide-routing
US20040191119A1 (en) * 2003-03-24 2004-09-30 Zanzucchi Peter J. Analyte concentration detection devices and methods
US20120196383A1 (en) * 2009-06-01 2012-08-02 Arthur Nitkowski Integrated optofluidic system using microspheres
US8675199B2 (en) * 2006-03-10 2014-03-18 Plc Diagnostics, Inc. Waveguide-based detection system with scanning light source
US20140178861A1 (en) * 2006-03-10 2014-06-26 Reuven Duer Waveguide-based detection system with scanning light source

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5544268A (en) * 1994-09-09 1996-08-06 Deacon Research Display panel with electrically-controlled waveguide-routing
US20040191119A1 (en) * 2003-03-24 2004-09-30 Zanzucchi Peter J. Analyte concentration detection devices and methods
US8675199B2 (en) * 2006-03-10 2014-03-18 Plc Diagnostics, Inc. Waveguide-based detection system with scanning light source
US20140178861A1 (en) * 2006-03-10 2014-06-26 Reuven Duer Waveguide-based detection system with scanning light source
US20120196383A1 (en) * 2009-06-01 2012-08-02 Arthur Nitkowski Integrated optofluidic system using microspheres

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10660619B2 (en) 2017-07-19 2020-05-26 Evanostics Llc Cartridges for oral fluid analysis and methods of use
US11262367B2 (en) 2017-12-15 2022-03-01 Evanostics Llc Optical reader for analyte testing
US11940454B2 (en) 2017-12-15 2024-03-26 Aspida Dx Inc. Optical reader for analyte testing
US11467086B2 (en) * 2019-08-30 2022-10-11 Canon Medical Systems Corporation Sample inspection apparatus

Similar Documents

Publication Publication Date Title
US10590493B2 (en) Waveguide-based detection system with scanning light source
US9423397B2 (en) Waveguide-based detection system with scanning light source
US8675199B2 (en) Waveguide-based detection system with scanning light source
US10551318B2 (en) Waveguide-based optical scanning systems
US11181479B2 (en) Waveguide-based detection system with scanning light source
US8187866B2 (en) Optical scanning system
US8288157B2 (en) Waveguide-based optical scanning systems
US8747751B2 (en) System and method for nucleic acids sequencing by phased synthesis
US7170050B2 (en) Apparatus and methods for optical analysis of molecules
US20130338013A1 (en) Substrates and optical systems and methods of use thereof
WO2010073605A1 (en) Fluorescence detector
WO2017059425A1 (en) Waveguide-based detection system with scanning light source
WO2015131056A2 (en) Waveguide-based detection system with scanning light source
JP5171668B2 (en) Method for correcting substrate misalignment

Legal Events

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

Ref document number: 16852824

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 16852824

Country of ref document: EP

Kind code of ref document: A1