CA2579150C - Apparatus and method for analysis of molecules - Google Patents

Apparatus and method for analysis of molecules Download PDF

Info

Publication number
CA2579150C
CA2579150C CA2579150A CA2579150A CA2579150C CA 2579150 C CA2579150 C CA 2579150C CA 2579150 A CA2579150 A CA 2579150A CA 2579150 A CA2579150 A CA 2579150A CA 2579150 C CA2579150 C CA 2579150C
Authority
CA
Canada
Prior art keywords
array
optical
confinements
nucleic acid
substrate
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CA2579150A
Other languages
French (fr)
Other versions
CA2579150A1 (en
Inventor
Stephen W. Turner
Jonas Korlach
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Pacific Biosciences of California Inc
Original Assignee
Pacific Biosciences of California Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US10/944,106 external-priority patent/US7170050B2/en
Application filed by Pacific Biosciences of California Inc filed Critical Pacific Biosciences of California Inc
Publication of CA2579150A1 publication Critical patent/CA2579150A1/en
Application granted granted Critical
Publication of CA2579150C publication Critical patent/CA2579150C/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

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
    • G01N21/6452Individual samples arranged in a regular 2D-array, e.g. multiwell plates
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L3/00Containers or dishes for laboratory use, e.g. laboratory glassware; Droppers
    • B01L3/50Containers for the purpose of retaining a material to be analysed, e.g. test tubes
    • B01L3/508Containers for the purpose of retaining a material to be analysed, e.g. test tubes rigid containers not provided for above
    • B01L3/5085Containers for the purpose of retaining a material to be analysed, e.g. test tubes rigid containers not provided for above for multiple samples, e.g. microtitration plates
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B82NANOTECHNOLOGY
    • B82YSPECIFIC USES OR APPLICATIONS OF NANOSTRUCTURES; MEASUREMENT OR ANALYSIS OF NANOSTRUCTURES; MANUFACTURE OR TREATMENT OF NANOSTRUCTURES
    • B82Y20/00Nanooptics, e.g. quantum optics or photonic crystals
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B82NANOTECHNOLOGY
    • B82YSPECIFIC USES OR APPLICATIONS OF NANOSTRUCTURES; MEASUREMENT OR ANALYSIS OF NANOSTRUCTURES; MANUFACTURE OR TREATMENT OF NANOSTRUCTURES
    • B82Y30/00Nanotechnology for materials or surface science, e.g. nanocomposites
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6869Methods for sequencing
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N35/00Automatic analysis not limited to methods or materials provided for in any single one of groups G01N1/00 - G01N33/00; Handling materials therefor
    • G01N35/02Automatic analysis not limited to methods or materials provided for in any single one of groups G01N1/00 - G01N33/00; Handling materials therefor using a plurality of sample containers moved by a conveyor system past one or more treatment or analysis stations
    • G01N35/028Automatic analysis not limited to methods or materials provided for in any single one of groups G01N1/00 - G01N33/00; Handling materials therefor using a plurality of sample containers moved by a conveyor system past one or more treatment or analysis stations having reaction cells in the form of microtitration 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
    • G02B6/10Light guides; Structural details of arrangements comprising light guides and other optical elements, e.g. couplings of the optical waveguide type
    • G02B6/12Light guides; Structural details of arrangements comprising light guides and other optical elements, e.g. couplings of the optical waveguide type of the integrated circuit kind
    • G02B6/13Integrated optical circuits characterised by the manufacturing method
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L2200/00Solutions for specific problems relating to chemical or physical laboratory apparatus
    • B01L2200/06Fluid handling related problems
    • B01L2200/0647Handling flowable solids, e.g. microscopic beads, cells, particles
    • B01L2200/0668Trapping microscopic beads
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L2300/00Additional constructional details
    • B01L2300/06Auxiliary integrated devices, integrated components
    • B01L2300/0627Sensor or part of a sensor is integrated
    • B01L2300/0654Lenses; Optical fibres
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L2300/00Additional constructional details
    • B01L2300/08Geometry, shape and general structure
    • B01L2300/0896Nanoscaled
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10TTECHNICAL SUBJECTS COVERED BY FORMER US CLASSIFICATION
    • Y10T436/00Chemistry: analytical and immunological testing
    • Y10T436/10Composition for standardization, calibration, simulation, stabilization, preparation or preservation; processes of use in preparation for chemical testing

Abstract

The present invention relates to optical confinements, method of preparing and methods of using them for analyzing molecules and/or monitoring chemical reactions. The apparatus and methods embodied in the present invention are particularly useful for high-throughput and low-cost single-molecular analysis.

Description

APPARATUS AND METHOD FOR ANALYSIS OF MOLECULES
BACKGROUND OF THE INVENTION
[0002] Confinement of illumination and signal detection has long been recognized as an important tool in molecular diagnostics since the application of Fluorescence Correlation Spectroscopy (FCS). FCS involves illumination of a sample volume containing fluorophore-labeled molecules, and detection of fluctuations in fluorescence signal produced by the molecules as they diffuse into and out of an effective observation volume. The fluorescence intensity fluctuations can best be analyzed if the volume under observation contains only a small number of fluorescent molecules, and if the background signal is low. This can be accomplished by the combination of a drastically limited detection volume and a low sample concentration. The detection volume of traditional FCS
is approximately 0.5 femtoliters (or 0.5 x le liters), and is achieved through the use of a high numerical aperture microscope objective lens to tightly focus a laser beam. In this detection volume, single molecules can be observed in solutions at concentrations of up to approximately one nanomolar. This concentration range is unacceptably low for most biochemical reactions, which have reaction constants that are typically in or above the micromolar range.
At lower concentrations, these reactions either do not proceed acceptably fast, or behave in a qualitatively different fashion than is useful in most analyses. To observe single molecules at higher, more relevant concentrations, the observation volume would typically need to be reduced to far smaller dimensions.
100031 In recent years, the advancement in nanofabrication technology enabled the production of nanoscale devices that are integrated with electrical, optical, chemical and/or mechanical elements.
100041 However, there still remains a considerable need for chemical and biological analyses that are faster, cheaper and of greater accuracy, to provide for the ability to observe single molecule reactions under conditions that are more biologically or diagnostically relevant. There also exists a need for small, mass produced, and disposable devices that can aid in these goals by providing optical confinements that are amenable to single-molecule analysis at a higher concentration. The present invention satisfies these needs and provides related advantages as well.

SUMMARY OF THE INVENTION
[0005] A principal aspect of the present invention is the design of optical devices and methods for characterizing molecules and/or monitoring chemical reactions. The devices and methods of the present invention are particularly =
suited for single-molecule analysis.
[0006] Accordingly, the present invention provides an array of optical confinements having a surface density exceeding 4 X104 confinements per mm2, preferably exceeding 105 confinements per mm2. In one aspect, the individual confinement in the array provide an effective observation volume that is less than one nanoliter (10X-9 liter), less than one picoliter, or less than one femtoliter, preferably on the order of zeptoliter. In other aspects, each of the individual confinement provides an effective observation volume that is less than 1000 zeptoliters, 100 zeptoliters, 80 zeptoliters, or less than 50 zeptoliters, or even less than 10 zeptoliters.
[0007] In other aspects, each of the individual confinement yields an effective observation volume that permits resolution of individual molecules present at a concentration that is higher than one nanomolar, or higher than 100 nanomolar, or on the order of micromolar range. In certain preferred aspects, each of the individual confinement yields an effective observation volume that permits resolution of individual molecules present at a physiologically relevant concentration, e.g., at a concentration higher than about 1 micromolar, or higher than 50 micromolar range or even higher than 100 micromolar. The array may comprise a zero-mode waveguide or other nanoscale optical structure. The array of optical confinements may further comprise another array of confinements that does not yield the above-described effective observation volume or does not permit resolution of individual molecules. For example, the array of optical confinements may be coupled with or integrated into a microtiter plate, where a separate array of optical confinements may be disposed within each of several different wells on a multiwell reaction plate. The array of optical confinement may comprise at least about 2X105 optical confinement, or at least about 106, or at least about 107 optical confinements.
[0008] In another embodiment, the present invention provides a method of creating a plurality of optical confinements having the aforementioned characteristics. The method involves the steps of (a) providing a substrate;
and (b) forming an array of optical confinements having a surface density exceeding 4 X104 confinements per mm2, wherein the individual confinement comprises a zero-mode waveguide comprising:
a cladding surrounding a core, wherein said cladding is configured to preclude propagation of electromagnetic energy of a wavelength longer than a cutoff wavelength longitudinally through the core of the zero-mode waveguide; and (c) illuminating the array with an electromagnetic radiation of a frequency less than the cutoff frequency, thereby creating the plurality of optical confinements.

[0009] In another embodiment, the present invention provides a method of creating an optical observation volume that permits resolution of individual molecules. The method involves providing a zero-mode waveguide that comprises a cladding surrounding a core, wherein said cladding is configured to preclude propagation of electromagnetic energy of a frequency less than a cutoff frequency longitudinally through the core of the zero-mode waveguide, wherein upon illuminating the zero-mode waveguide with an electromagnetic radiation of a frequency less than the cutoff frequency, the zero-mode waveguide yields an effective observation volume that permits resolution of individual molecules. In certain aspects, the effective observation volume is less than one nanoliter (10-9 liter), less than one picoliter, or less than one femtoliter, preferably on the order of zeptoliters. Using the zero-mode waveguide of the present invention, one typically can obtain an effective observation volume that is less than 100 zeptoliter (100 x 10-21 liters) or less than 50 zeptoliters, or even less than 10 zeptoliters. In other aspects, the method yields an effective observation volume that permits resolution of individual molecules present at a concentration that is higher than one nanomolar, more often higher than 100 nanomolar, and preferably on the order of micromolar range. In preferred embodiments, individual molecules present at a concentration higher than about 5 micromolar, or higher than 7.5 micromolar, or even higher than 50 micromolar range, can be resolved by the method of the present invention.
[0010] The present invention also provides a method of detecting interactions among a plurality of molecules. The method comprises the steps of (a) placing the plurality of molecules in close proximity to an array of zero-mode waveguides, wherein individual waveguides in the array are separated by a distance sufficient to yield detectable intensities of diffractive scattering at multiple diffracted orders upon illuminating the array with an incident wavelength; (b) illuminating the array of zero-mode waveguides with an incident wavelength; and (c) detecting a change in the intensities of diffractive scattering of the incident wavelength at the multiple diffracted orders, thereby detecting the interactions among a plurality of molecules.
[0011] The present invention also provides a method of reducing diffractive scattering upon illuminating an array of optical confinements with an incident wavelength, wherein the array comprises at least a first optical confinement and a second optical confinement, said method comprising: forming the array of optical confinements wherein the optical confinement is separated from the second optical confinement by a distance such that upon illumination with the incident wavelength, intensity of diffractive scattering resulting from the first optical confinement at a given angle is less than that if the first optical confinement were illuminated with the same incident wavelength in the absence of the optical confinement. In preferred aspects, the aforementioned optical confinements are zero mode waveguides.
[0012] The present invention also includes a method of detecting a biological analyte using an array of optical confinements having a density on a substrate exceeding 4X104 confinements per mm2 or any other density described herein or equivalents thereof The method typically involves illuminating at least one optical confinement within the array that is suspected to contain the analyte with an incident light beam.
The invention also provides a method of using of an array of optical confinements having a density on a substrate exceeding 4X104 confinements per mm2 any other density described herein or equivalents thereof for performing multiple chemical reactions. The method comprises the steps of placing the plurality of reaction samples comprising labeled reactants into the optical confinements in the array, wherein a separate reaction sample is placed into a different confinement in the array;
subjecting the array to conditions suitable for formation of products of the chemical reactions; and detecting the formation of the products with said optical system.
[0013] In addition, the invention provides a method of sequencing a plurality of target nucleic acid molecules. The method typically involves (a) providing an array of optical confinements having a density on a substrate exceeding 4X104 confinements per mm2, or any other density described herein or equivalents thereof, wherein said optical confinements provide an effective observation volume that permits observation of individual molecules; and an optical system operatively coupled to the optical confinements that detects signals from the effective observation volume of said confinement; (b) mixing in the optical confinements the plurality of target nucleic acid molecules, primers complementary to the target nucleic acid molecules, polymerization enzymes, and more than one type of nucleotides or nucleotide analogs to be incorporated into a plurality of nascent nucleotide strands, each strand being complementary to a respective target nucleic acid molecule; (c) subjecting the mixture of step (b) to a polymerization reaction under conditions suitable for formation of the nascent nucleotide strands by template-directed polymerization of the nucleotides or nucleotide analogs; (d) illuminating the optical confinements with an incident light beam; and (e) identifying the nucleotides or the nucleotide analogs incorporated into the each nascent nucleotide strand.
[0014] The present invention also provides an apparatus comprising an array of waveguides on a solid support having a fill fraction greater than about 0.0001, wherein said waveguides are suitable for holding a biological reagent, and wherein waveguides provide an effective observation volume that permits observation of individual molecules present in said biological reagent; and an optical system that detects said individual molecules in said waveguides, by e.g., detecting signals from the effective observation volume.
In one aspect, the array has a fill fraction greater than about 0.001. In another aspect, the array has a fill fraction greater than about 0.01, in some instances greater than 0.1, or within the range about 0.001 to about 0.1.
[0015] The present invention also provides various methods of using such high fill fraction array. In one embodiment, the present invention provides a method of detecting a biological analyte. The method comprises optically capturing the analyte within an optical confinement that is created by (a) providing an array of waveguides having a fill fraction greater than about 0.0001; and (b) illuminating at least one waveguide within the array that is suspected to contain the analyte with an incident light beam thereby detecting the analyte.
[00161 In another embodiment, the present invention provides a method of performing multiple chemical reactions involving a plurality of reaction samples using the subject high fill fraction array. The method involves (a) providing a subject high fill fraction array; (b) placing the plurality of reaction samples comprising labeled reactants into the waveguides in the array, wherein a separate reaction sample is placed into a different waveguide in the array; (c) subjecting the array to conditions suitable for formation of products of the chemical reactions; and (d) detecting the formation of the products with an optical system. The step of detecting may comprise illuminating the different waveguides with an incident light beam and detecting an optical signal emitted from the reaction samples.
Applicable chemical reactions may involve protein-protein interactions, nucleic acid-protein interactions, and nucleic acid-nucleic acid interactions. Specifically, the present invention provides a method of sequencing a plurality of target nucleic acid molecule using a fill fraction greater than about 0.0001.
[0017] The present invention further provides a method of sequencing nucleic acid using an array having a high fill faction. The method typically involves a) providing an array of waveguides having a fill fraction greater than about 0.0001, or 0.001, or 0.01 or even 0.1; (b) mixing in the waveguides the plurality of target nucleic acid molecules, primers complementary to the target nucleic acid molecules, polymerization enzymes, and more than one type of nucleotides or nucleotide analogs to be incorporated into a plurality of nascent nucleotide strands, each strand being complementary to a respective target nucleic acid molecule; (c) subjecting the mixture of step (b) to a polymerization reaction under conditions suitable for formation of the nascent nucleotide strands by template-directed polymerization of the nucleotides or nucleotide analogs; (d) illuminating the waveguides with an incident light beam; and (e) identifying the nucleotides or the nucleotide analogs incorporated into the each nascent nucleotide strand.
[0018] Also included in the present invention is a redundant sequencing method. The method comprises (a) subjecting a target nucleic acid molecule to a template-directed polymerization reaction to yield a nascent nucleic acid strand that is complementary to the target nucleic acid molecule in the presence of a plurality of types of nucleotides or nucleotide analogs, and a polymerization enzyme exhibiting strand-displacement activity; and (b) registering a time sequence of incorporation of nucleotides or nucleotide analogs into the nascent nucleotide strand.
In one aspect of this embodiment, the target nucleic acid molecule is a circular nucleic acid, or is a linear or circular template strand synthesized from a circular nucleic acid sequence such that the synthesized strand includes multiple repeated copies of the original circular strand, and is thus is subject to the sequencing operations of the invention. In another aspect of this embodiment, the target nucleic acid molecule is sequenced multiple times, e.g., more than once, or more than twice by the polymerization enzyme. In yet another aspect of this embodiment, the polymerization enzyme is a DNA polymerase, such as a modified or unmodified 4329 polymerase.
[0019] Further included in the present invention is a solid support having a surface wherein the surface has a polymerization enzyme array attached to it, wherein members of the array comprise individually and optically resolved polymerization enzymes possessing strand-displacement activities.
[0020] Also provided is a zero mode waveguide, comprising a first molecular complex immobilized therein, said molecular complex comprising a polymerization enzyme complexed with a target nucleic acid, wherein the polymerization enzyme processes a sequence of nucleotides in said target nucleic acid multiple times via template-dependent replication of the target nucleic acid.
[0021] Further provided by the present invention is a method of fabricating an array of optical confinements that exhibits a minimal intensity of diffractive scattering of an incident wavelength. The method comprises providing a substrate; and forming the array of optical confinements on the substrate such that individual confinements in the array are separated from each other at a distance less than one half of the wavelength.
[0022] Finally, the present invention includes a method of fabricating an optical confinement the method comprises a cladding surrounding a core, comprising: (a) providing a substrate coated with a layer of photoresist; (b) patterning said layer of photoresist to define boundaries of said core; (c) removing said layer of photoresist surrounding said defined boundaries so that a sufficient amount of photoresist remains to occupy said core; (d) depositing a layer of cladding material over said remaining photoresist and said substrate; (e) removing at least a = portion of said cladding material deposited over said remaining photoresist; and (f) removing said photoresist of step (e) to form said core surrounded by said cladding of said optical confinement.
In one aspect, the photoresist is negative and said patterning step employs a positive pattern. In another aspect, the photoresist is positive and said patterning step employs a negative pattern. The removing step can be effected by a technique selected from the group consisting of etching, mechanical polishing, ion milling, and solvent dissolution. The layer of cladding material can be deposited by a thermal evaporation method or vapor deposition.
BRIEF DESCRIPTION OF THE DRAWINGS
[0023] Figure 1 depicts a top view of an array of illustrative optical confinements, here zero-mode waveguides arranged in a square format.
[0024] Figure 2 depicts a top view of an array of illustrative optical confinements, here zero-mode waveguides arranged in a non-square format.
[0025] Figure 3 depicts a top view of an illustrative 2-dimentional array with an illustrative angle and two different unit vector lengths.
[0026] Figure 4 depicts a top view of an illustrative regular disposition of ZMWs.
[0027] Figure 5 depicts an array of arrays, in which a subarray 71 is part of a super array 72.
[0028] Figure 6 illustrates a process of negative tone fabrication.
100291 Figure 7 illustrates an array of ZMWs optically linked to an optical system.
[0030] Figure 8 depicts a scanning electron micrographs of ZMW structures fabricated by positive tone resist (left panels) or negative tone resist (right panels). The grain structure of the polycrystalline film is visible in the image as flecks, and the ZMWs as dark round structures.
[0031] Figure 9 depicts a single-molecule DNA sequence pattern recognition in ZMWs using artificial pre-formed replication forks.
[0032] Figure 10, depicts a coated ZMW 101 that is bound to a substrate 105.
The ZMW comprises a sidewall 102, a coating 103 on the upper surface, and a metal film 104.
[0033] Figure 11 depicts one alignment strategy and optical setup.
[0034] Figure 12 depicts an alternative optical confinement made of porous film 91 on a substrate 93. 92 represents the pores in the film.
[0035] Figures 13A-B depict an alignment detection system and the associated components.
[0036] Figure 14 depicts several exemplary photocleavable blockers and the applicable wavelength applied to cleave the blocking groups.
[0037] Figure 15 depicts an exemplary reversible extension terminator in which the photocleavable blocker is conjugated to a detectable label (e.g., fluorescent label).
[0038] Figure 16 depicts an exemplary profile of fluorescent bursts corresponding to the time sequence of incorporation of two types of labeled nucleotides or nucleotide analogs in single-molecule sequencing reaction using the subject optical confinement.
DETAILED DESCRIPTION OF THE INVENTION
[0039] The practice of the present invention will employ, unless otherwise indicated, conventional techniques of Integrated Circuit (IC) processing biochemistry, chemistry, molecular biology, genornics and recombinant DNA, which are within the skill of the art. See, e.g., Stanley Wolf et al., SILICON
PROCESSING FOR THE VLSI ERA, Vols 1-4 (Lattice Press); Michael Quirk et al., SEMICONDUCTOR MANUFACTURING
TECHNOLOGY;
Sambrook, Fritsch and Maniatis, MOLECULAR CLONING: A LABORATORY MANUAL, 2"
edition(1989); the series METHODS IN ENZYMOLOGY (Academic Press, Inc.): PCR 2: A PRACTICAL
APPROACH (Mi.
MacPherson, B.D. Hames and G.R. Taylor eds. (1995).
Definitions:
100401 As used in the specification and claims, the singular form "a", "an"
and "the" include plural references unless the context clearly dictates otherwise.
[0041] "Luminescence" refers 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 then move from an "excited state" to a lower energy state (usually the ground state);
this process is often referred to as "decay".
There are many causes of excitation. If 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" or "luminescent signal"
may have any one of the above-described properties.

100421 The term "electromagnetic radiation" refers to electromagnetic waves of energy including, for example, in an ascending order of frequency (or alternatively, in a descending order of wavelength), infrared radiation, visible light, ultraviolet (UV) light, X-rays, and gamma rays.
100431 As used herein, an "effective observation volume" typically refers to that volume that is observable by the detection means employed for a given application. For example, in the case of fluorescence based detection, it is that volume which is exposed to excitation radiation and/or from which emission radiation is gathered by an adjacent optical train/detector. By way of example, in the case of a zero mode waveguide used for certain applications, an effective observation volume is dictated by the propagation of excitation radiation into the waveguide core, and particularly that volume that is exposed to light that is at least 1%, and preferably at least 10%
of the original intensity of excitation radiation entering the waveguide core.
Such intensities and volumes are readily calculable from the particular conditions of the application in question, including the wavelength of the excitation radiation and the dimensions of the waveguide core (See, e.g., U.S.
Patent No. 6,917,726).
100441 A "primer" is a short polynucleotide, generally with a free 3' OH
group, that binds to a target nucleic acid (or template) potentially present in a sample of interest by hybridizing with the target nucleic acid, and thereafter promoting polymerization of a polynucleotide complementary to the target.
100451 The terms "operatively linked to" or "operatively coupled to" are used interchangeably herein. They refer to a juxtaposition wherein the components so described are in a relationship permitting them to function in their intended manner.
100461 The term "nucleotide" generally refers to a molecule comprising a base, sugar and one or more anionic groups, preferably phosphates. The molecule may comprise one, two, three, four, five or more phosphates groups and/or other groups such as sulfate. The term also encompasses nucleotide analogs that are structurally analogous to naturally occurring nucleotides and are capable of acting substantially like nucleotides, for example exhibiting base complementarity with one or more of the bases that occur in DNA or RNA, and/or being capable of base-complementary incorporation in synthesizing nucleotide strand by a polymerization enzyme.
100471 A "type of nucleotide" refers to a set of nucleotides that share a common characteristic that is to be detected. For instance, the types of nucleotides can be classified into four categories: A, T, C, and G for DNA, or A, U, C and G for RNA. In some embodiments, each type of nucleotides used in the a reaction will be labeled with a unique label that is distinguishable from the rest.
100481 The term "polynucleotide" refers to a polymeric form of "nucleotides"
of any length.
[0049] The term "optical confinement" refers to an area in which the reactants for an intended reaction within the confinement are confined and resolved by optical means.
100501 A "polynucleotide probe" refers to a polynucleotide used for detecting or identifying its corresponding target polynucleotide in a hybridization reaction.
[0051] The term "hybridize" as applied to a polynucleotide refers to the ability of the polynucleotide to form a complex that is stabilized via hydrogen bonding between the bases of the nucleotide residues in a hybridization reaction. The hydrogen bonding may occur by Watson-Crick base pairing, Hoogstein binding, or in any other sequence specific manner. The complex may comprise two strands forming a duplex structure, three or more strands forming a multi-stranded complex, a single self-hybridizing strand, or any combination of these. The hybridization reaction may constitute a step in a more extensive process, such as the initiation of a PCR reaction, or the enzymatic cleavage of a polynucleotide by a ribozyme.
[0052] Hybridization can be performed under conditions of different "stringency". Relevant conditions include temperature, ionic strength, time of incubation, the presence of additional solutes in the reaction mixture such as formamide, and the washing procedure. Higher stringency conditions are those conditions, such as higher temperature and lower sodium ion concentration, which require higher minimum complementarity between hybridizing elements for a stable hybridization complex to form. In general, a low stringency hybridization reaction is carried out at about 40 C in 10 x SSC or a solution of equivalent ionic strength/temperature. A moderate stringency hybridization is typically performed at about 50 C in 6 x SSC, and a high stringency hybridization reaction is generally performed at about 60 C in 1 x SSC.
[0053] When hybridization occurs in an antiparallel configuration between two single stranded polynucleotides, the reaction is called "annealing" and those polynucleotides are described as "complementary". A double-stranded polynucleotide can be "complementary" or "homologous" to another polynucleotide, if hybridization can occur between one of the strands of the first polynucleotide and the second.
"Complementarity" or "homology" (the degree that one polynucleotide is complementary with another) is quantifiable in terms of the proportion of bases in opposing strands that are expected to form hydrogen bonding with each other, according to generally accepted base pairing rules.

Structure of the Optical Confinements of the Present Invention [0054] One aspect of the present invention is the design of optical devices and methods for characterizing molecules and/or monitoring chemical reactions. The optical devices of the present invention allow multiplexing of large numbers of single-molecule analyses under physiologically relevant conditions.
[00551 In one embodiment, the present invention provides a high density array of optical confinements having a surface density exceeding 4 X104 confinements per min2, preferably exceeding 105, wherein the individual confinement in the array provides an effective observation volume on the order of zeptoliters. The array may contain at least about 2 X 105, at least about 106, or at least about 107 optical confinements. Preferably, the individual confinement in the array provides an effective observation volume less than about 1000 zeptoliters, more preferably less than about 900, more preferably less than about 80, even more preferably less than about 10 zeptoliters. Where desired, an effective observation volume less than 1 zeptoliter can be provided. In a preferred aspect, the individual confinement yields an effective observation volume that permits resolution of individual molecules present at a physiologically relevant concentration. The physiologically relevant concentrations for most biochemical reactions range from micro-molar to millimolar because most of the enzymes have their Michaelis constants in these ranges. Accordingly, preferred array of optical confinements has an effective observation volume for detecting individual molecules present at a concentration higher than about 1 micromolar ( M), or more preferably higher than 50 M, or even higher than 100 M.
[0056] To achieve the required observation volume for single-molecule analysis under physiologically relevant conditions, the array may comprise zero-mode waveguides or alternative nanoscale optical structures. Such alternative structures include but are not limited to porous films with reflective index media, and confinements using index matching solids.
[0057] As used herein, "zero-mode waveguide" refers to an optical guide in which the majority of incident radiation is attenuated, preferably more than 80%, more preferably more than 90%, even more preferably more than 99% of the incident radiation is attenuated. As such high level of attenuation, no significant propagating modes of electromagnetic radiation exist in the guide. Consequently, the rapid decay of incident electromagnetic radiation at the entrance of such guide provides an extremely small observation volume effective to detect single molecules, even when they are present at a concentration as high as in the micromolar range.
[0058] The zero-mode waveguide of the present invention typically comprises a cladding surrounding a core (i.e., partially or fully), wherein the cladding is configured to preclude propagation of electromagnetic energy of a wavelength higher than the cutoff wavelength longitudinally through the core of the zero-mode waveguide. The cladding is typically made of materials that prevent any significant penetration of the electric and the magnetic fields of an electromagnetic radiation. Suitable materials for fabricating the cladding include but are not limited to alloys, metals, and semi-conducting materials, and any combination thereof. Alloys include any of the numerous substances having metallic properties but comprising two or more elements of which at lest one is a metal. Alloys may vary in the content or the amount of the respective elements-whether metallic or non metallic. Preferred alloys generally improve some desirable characteristics of the material over a pure elemental material. Characteristics that can be improved through the use of mixtures of materials include, chemical resistance, thermal conductivity, electrical conductivity, reflectivity, grain size, coefficient of thermal expansion, brittleness, temperature tolerance, conductivity, and/or reduce grain size of the cladding.
100591 In general, alloys suitable for the present invention may involve mixtures where one component is present at fractions as low as 0.0001%. In other instances, alloys with large fractions of more than one compound will be desirable. One embodiment of the ZMW uses aluminum as the cladding of the ZMW
structure. As an example of how alloys can be beneficial to a ZMW structure, it is useful to consider different alloys of aluminum in how they would affect a ZMW. In the art of metalurgy, numerous materials are alloyed with aluminum. Non-limiting examples of materials suitable to alloy with aluminum are antimony, arsenic, beryllium, bismuth, boron, cadmium, calcium, carbon, cerium, chromium, cobalt, copper, gallium, hydrogen, indium, iron, lead, lithium, magnesium, manganese, mercury, molybdenum, nickel, niobium, phosphorous, silicon, vanadium, zinc and others. By way of example of how the introduction of another element could beneficially impact the ZMW performance, the introduction of boron to aluminum is known to increase the conductivity of aluminum. An increase in conductivity of the metal film may improve the performance by decreasing the penetration depth thereby decreasing the observation volume. A preferred embodiment includes an alloy of aluminum that is more than 0.0001% of a dopant.
A more preferred embodiment includes an alloy of aluminum that is more than 0.005% of a dopant. A still more preferred embodiment includes an allow of aluminum that is more than 0.1% of a dopant.
[0060] In contrast, some materials are expected to decrease the performance of the ZMW structure, and in these instances it will be desirable to take measures to eliminate certain impurities. For example, in certain applications it may be desirable to decrease the amount of lead or arsenic if toxicity of the device is a concern. A preferred embodiment of the device includes a metal film that is less than 1% arsenic. A
more preferred embodiment of the device includes a metal films that is less than 0.1% arsenic. A still more preferred embodiment includes a metal film that is less than 0.001% arsenic. A still more preferred embodiment includes a metal film that is less than 0.00001% arsenic. An additional preferred embodiment includes a metal film that is less than 1% lead. A still more preferred embodiment includes a metal film that is less than 0.1% lead. A
still more preferred embodiment includes a metal film that is less than 0.01% lead. A still more preferred embodiment includes a metal film that is less than 0.001% lead. A still more preferred embodiment includes a film that is less than 0.00001% lead. In other applications where optical confinement performance is especially important, impurities that tend to reduce the conductivity, thereby worsening the confinement, will be undesirable. For example, vanadium is known in the art of metallurgy to reduce the conductivity of aluminum. A preferred embodiment includes a metal film that is less than 0.1% vanadium. A still more preferred embodiment includes a metal film that is less than 0.01% vanadium. A still more preferred embodiment includes a film that is less than 0.001% vanadium.
[0061] Semi-conducting materials suitable for fabricating the cladding are generally opaque, and they include silicon, silicates, silicon nitride, gallium phosphide, gallium arsenide, or any combinations thereof.
[0062] The cladding of the subject zero-mode waveguide may be coated with materials to improve the surface quality. For instance, coating may enhance the durability of the cladding material. In addition, coating is particularly desirable if the reactants contained in the core are prone to interact or adhere to the cladding material. A
variety of appropriate coating materials are available in the art. Some of the materials may covalently adhere to the surface, others may attach to the surface via non-covalent interactions. Non-limiting examples of coating materials include aluminum oxide film, silanization reagent such as dimethychlorosilane, dimethydichlorosilane, hexamethyldisilazane or trimethylchlorosilane, polymaleimide, and siliconizing reagents such as silicon oxide, AquasilTM, and SurfasilTm. An illustrative coated ZMW (101) is shown in Figure 10. The ZMW (101) is bound to a substrate 105. The ZMW comprises a sidewall 102, a coating 103 on the upper surface, and a metal film 104.
[0063] In certain embodiments, it may be advantageous to construct the confinement from metal compositions that are inhomogeneous combinations of more than one material. For example, for certain applications, it may be beneficial to provide a composition that comprises more than one layer, each layer having a different composition, or composition that varies within a layer. This can have beneficial effects on several aspects of the performance of the confinement, including but not limited to the nature of the optical confinement, the structural strength and behavior of the device, the characteristics of the surface chemistry of the device or the like. In one embodiment the confinement comprises two layers in which one of the layers serves to enhance the adhesion of the second layer to a substrate. In another embodiment, the composition of the cladding film varies as a function of the axial position relative to the confinement, so as to provide different optical performance than would be obtained from a layer of uniform composition. In a particular version of this embodiment, the film comprises a composition that has a larger value of skin depth close to the surface of the substrate, and comprises a composition that has a smaller value of skin depth farther from the surface of the substrate, so that the nature of the confinement is to be more uniform in shape near the surface and then tapering off more quickly a larger distances away from the substrate. In another embodiment, the thicknesses of two different layers comprising the cladding of the confinement are chosen so that a specific optical condition is achieved at the substrate of the device, such as constructive or destructive interference.
[0064] The internal cavity (i.e., the core) surrounded by the cladding may adopt a convenient size, shape or volume so long as propagating modes of electromagnetic radiation in the guide is effectively prevented. The core typically has a lateral dimension less than the cutoff wavelength (k). For a circular guide of diameter d and having a clad of perfect conductor, xe is approximately 1.7 X d. The cross sectional area of the core may be circular, elliptical, oval, conical, rectangular, triangular, polyhedral, or in any other shape. The various shapes can have particular suitability for certain applications. For instance, elongated cross-sections can be useful to provide enhanced access to molecules with mechanical persistence or stiffness, such as DNA. Cross sections ranging from extended slots to ovals of various aspect ratio will significant increase the accessibility of the persistent molecule to the detection zone of the structure, without excessive compromise in the axial attenuation of radiation. Although uniform cross sectional area is preferred, the cross sectional area may vary at any given depth of the guide if desired.
Preferred average cross sectional areas range from 100 nm2 to 10,000 nm2.
[0065] In a preferred embodiment, the core is non-cylindrical. In one aspect of this embodiment, a non-cylindrical core comprises an opening on the upper surface and a base at the bottom surface that is entirely surrounded by the cladding, wherein the opening is narrower in lateral dimension than the base.
This configuration significantly restricts the diffusion of reactants, and hence increases the average residence time in the observation volume. Such configuration is particularly useful for measuring the association rate constant (on-rate) of a chemical reaction. In another aspect, the core comprises an opening that is wider in lateral dimension than the base. Such configuration allows easier access to large molecules that impose a steric or entropic hindrance to entering the structure if the open end of the zero mode waveguide was as small as the base needed to be for optical performance reasons. Examples include the accessibility for long strand polyelectrolytes such as DNA
molecules that are subject to entropic forces opposing entry into small openings.
[0066] The zero-mode waveguides embodied in the present invention have a relatively high fill fraction ratio, typically above 0.0001, preferably above 0.001, more preferably above 0.01, and even more preferably above 0.1.
As used herein, "fill fraction" of a pattern refers to the ratio of the area occupied by the foreground of the pattern to the total area occupied by the pattern (foreground and background, together).
The terms "fill fraction ratio" and "fill faction" are used interchangeably. In the context of zero-mode waveguide, the foreground is considered to be the area occupied by the core of the zero-mode waveguide, and the background is the area between the zero-mode waveguide (e.g., the aluminum film that forms the cladding in certain designs). The zero-mode waveguides with high fill fraction ratios are particularly useful for performing homogenous assays. The fill fraction can be calculated by summing the total areas of all of the zero-mode waveguides in the array and dividing by the total available area including both the zero-mode waveguides and the spaces between them. For example, if a zero-mode waveguide has a diameter of 50 nm, then the area of this zero-mode waveguide is one fourth of 7,850 square nanometers or 1962.5 nm2. If these zero-mode waveguides are in a square array separated by 100 nm, the total available area is 10,000 square nanometers for each zero-mode waveguide. Therefore, the array has a fill fraction of one fourth of 78% or 19.6%, which would provide nearly four orders of magnitude higher signal strength in a surface binding assay than a zero-mode waveguide having a fill fraction on the order of 0.01%.
[0067] In a bioassay such as an ELISA or other molecular binding bioassay, one limitation is the inability to operate "homogeneously", or in a mode where solutions may be added to a mixture but nothing removed. This complicates highly multiplexed assays, as provisions for both adding and removing material from a large number of wells is significantly more complex than the provisions for simply adding materials. In the case of the ELISA assay, the removal of materials is necessary, because the fluorescent (or other) markers that remain free in solution at the end of the assay would interfere with the ability to detect markers bound to the reaction surface. Techniques to overcome this have been devised to exploit the short range of radioactive emissions from certain radioisotopes, but these techniques have inherent difficulties associated with personnel safety and waste disposal. Other methods for confining the sensitivity of the assay to the surface have been devised, such as total internal reflection confinement (TIR), and confocal detection. The zero-mode waveguide photonic structure allows a simpler and less expensive optical system configuration than either of these techniques, and vastly outperforms both from the perspective of confinement of sensitivity to the surface.
[0068] The fill fraction is important in bioassays, because the effective probe area is limited to the surface area of the bottoms of the zero-mode waveguide in the detection region. The amount of signal detectable in such an assay will be directly proportional to the available area, and having a larger fraction of the available surface occupied by zero-mode waveguides will thus increase the signal strength of measurements of such assays. A high fill fraction structure would be generally useful in any surface sensitivity application, not limited to the ELISA assay.
[0069] The cutoff wavelength is the wavelength above which the waveguide is essentially incapable of propagating electromagnetic energy along the waveguide under the illumination geometry used. Given the geometry of the core, and the properties of the cladding material, as well as the wavelength of the incident electromagnetic radiation, one skilled in the art can readily derive the cutoff wavelength by solving the Maxwell's equations (see, e.g., John D. Jackson, CLASSICAL ELECTRODYNAMICS, second edition, John Willey and Sons). The choice of the incident wavelength will depend on the particular application in which the subject array is to be employed. In certain aspects, the incident wavelength may be selected from a range of about 10 nm to about 1 mm. For detecting fluorescent signals, the incident wavelength is typically selected from the range of about 380 tun to about 800 nm. Polarized (linearly or preferably circularly polarized) or unpolarized incident radiation is generally employed to illuminate the array in order to create a desired observation volume.
[0070] In a separate embodiment, the present invention provides an alternative optical confinement termed external reflection confinement (ERC). In contrast to the conventional total internal reflection confinement (IRC), the low index medium is the electromagnetic radiation carrier, and the high index (and opaque) medium is the reflector. As such, the roles of the refractive indices are reversed as compared to the IRC situation. ERC generally requires some kind of means to provide the analyte (i.e., the molecules under investigation) in the opaque phase.
[0071] IRC relies on reflection of an electromagnetic radiation incident on an interface between high index of refraction and low index of refraction. When light is incident above the critical angle of total internal reflection (known in the art), all of the incident electromagnetic radiation is reflected and none is transmitted into the low index phase. A thin region of evanescent radiation is established proximal to the interface on the low index side.
This radiation field is typically an exponentially decaying field with an attenuation length in the range from about 100 nm to about 200 nm, depending on the angle of incidence and the indices of refraction of the two phases. If the low index phase is a solution containing an analyte, then the evanescent radiation can be used to probe the analyte in the solution with a high degree of surface sensitivity.
[0072] In ERC, the carrier of the propagating electromagnetic radiation is a transparent low index film, and the analyte-bearing medium is a high-index metallic opaque film. In this case, most of the radiation is reflected irrespective of the angle of incidence, and non-reflected light is rapidly attenuated according to the skin depth of the metal. Typically, means is provided to convey the analyte within the metal phase. Theses means can take the form of a nanocapillary tube constructed within the metal layer. When sufficiently small, the presence of such a tube will have little effect on the distribution of energy in the two media, but can be amply large enough to convey biomolecules. To be small enough, any defects in the metal film must be small compared with the wavelength of the illumination. This can be achieved because of the large ratio between the wavelength of visible light, and the typical size of biomolecules of interest. While visible light is typically between 400 nm and 750 nm in wavelength, biomolecules of interest are generally in the vicinity of 1-30 nm in diameter.
The attenuation of the radiation at the interface can be used to confine illumination to a very small region of the analyte. A small hole in an index matched (to water) film on a high index substrate could provide lateral confinement beyond what is possible with diffraction limited optics in the TIR context. This could give 100 zeptoliter confinement in principle. In this method, a version of total internal reflection confinement is used in which a solid material index-matched to the analyte solution is applied to the substrate surface and then perforated with nanoscale holes.
When used in TIR mode, these structures will provide additional confinements above what can be obtained with TIR
alone.
[0073] Other alternative confinements are index matching solids. As an illustrative example, such optical confinement can be fabricated starting with a high index transparent susbtrate such as sapphire, spin coat 200 nm of PMMA (polymethyl methacrylate) resist resin. Exposure to electron beam lithography will render isolated spots soluble according to the pattern applied. After development, the device will have nano-scale holes in the PMMA
layer and are ready to be used in a TIR setup. Axial confinement is unaffected by the PMMA layer, as it has nearly the same index of refraction as the solution containing the analyte, but the solution is physically prevented from approaching near the surface except where the holes are situated, providing a degree of lateral confinement given by the diameter of the holes.
100741 The optical confinements can be provided with an optical system capable of detecting and/or monitoring interactions between reactants at the single-molecule level. Such optical system achieves these functions by first generating and transmitting an incident wavelength to the reactants contained in the confinements, followed by collecting and analyzing the optical signals from the reactants. Such systems typically employ an optical train that directs signals from an array of confinements onto different locations of an array-based detector to simultaneously detect multiple different optical signals from each of multiple different confinements. In particular, the optical trains typically include optical gratings or wedge prisms to simultaneously direct and separate signals having differing spectral characteristics from each confinement in an array to different locations on an array based detector, e.g., a CCD. By separately directing signals from each confinement to different locations on a detector, and additionally separating the component signals from each confinement to separate locations, one can simultaneously monitor multiple confinements, and multiple signals from each confinement.
[0075] The optical system applicable for the present invention comprises at least two elements, namely an excitation source and a photon detector. The excitation source generates and transmits incident light used to optically excite the reactants contained in the optical confinement. Depending on the intended application, the source of the incident light can be a laser, laser diode, a light-emitting diode (LED), a ultra-violet light bulb, and/or a white light source. Where desired, more than one source can be employed simultaneously. The use of multiple sources is particularly desirable in applications that employ multiple different reagent compounds having differing excitation spectra, consequently allowing detection of more than one fluorescent signal to track the interactions of more than one or one type of molecules simultaneously. A wide variety of photon detectors are available in the art.
Representative detectors include but are not limited to optical reader, high-efficiency photon detection system, photodiode (e.g. avalanche photo diodes (APD)), camera, charge couple device (CCD), electron-multiplying charge-coupled device (EMCCD), intensified charge coupled device (ICCD), and confocal microscope equipped with any of the foregoing detectors. Where desired, the subject arrays of optical confinements contain various alignment aides or keys to facilitate a proper spatial placement of the optical confinement and the excitation sources, the photon detectors, or the optical transmission element as described below.
[0076] The subject optical system may also include an optical transmission element whose function can be manifold. First, it collects and/or directs the incident wavelength to the optical confinement containing the reactants.
Second, it transmits and/or directs the optical signals emitted from the reactants inside the optical confinement to the photon detector. Third, it may select and/or modify the optical properties of the incident wavelengths or the emitted wavelengths from the reactants. Illustrative examples of such element are diffraction gratings, arrayed waveguide gratings (AWG), optic fibers, optical switches, mirrors, lenses (including microlens and nanolens), collimators.
Other examples include optical attenuators, polarization filters (e.g., dichroic filter), wavelength filters (low-pass, band-pass, or high-pass), wave-plates, and delay lines. In some embodiments, the optical transmission element can be planar waveguides in optical communication with the arrayed optical confinements. For instance, a planar waveguides can be operatively coupled to an array of zero-mode waveguides to directly channel incident wavelengths to the respective cores of the zero-mode waveguides so as to minimize the loss of wave energy. The planar channel can be included as a detachable unit located at the base of array substrate, or it can be bonded to the substrate as an integral part of the array.
[0077] The optical transmission element suitable for use in the present invention encompasses a variety of optical devices that channel light from one location to another in either an altered or unaltered state. Non-limiting examples of such optical transmission devices include optical fibers, diffraction gratings, arrayed waveguide gratings (AWG), optical switches, mirrors, (including dichroic mirrors), lenses (including microlens and nanolens), collimators, filters, prisms, and any other devices that guide the transmission of light through proper refractive indices and geometries.
[0078] In a preferred embodiment, the optical confinement of the present invention is operatively coupled to a photon detector. For instance, the arrayed optical confinement is operatively coupled to a respective and separate photon detector. 'The confinement and the respective detector can be spatially aligned (e.g., 1:1 mapping) to permit an efficient collection of optical signals from the waveguide. A particularly preferred setup comprises an array of zero-mode waveguides, wherein each of the individual waveguides is operatively coupled to a respective microlens or a nanolens, preferably spatially aligned to optimize the signal collection efficiency. Alternatively, a combination of an objective lens, a spectral filter set or prism for resolving signals of different wavelengths, and an imaging lens can be used in an optical train, to direct optical signals from each confinement to an array detector, e.g., a CCD, and concurrently separate signals from each different confinement into multiple constituent signal elements, e.g., different wavelength spectra, that correspond to different reaction events occurring within each confinement.
[0079] An exemplary optical setup is shown in Figure 7, in which an array of ZMWs is optically linked to an optical system. This system comprises a ZMW array film (81), a glass cover slip (82) through which light transmits and further converges through set of integral lenses (83) made of a material having a different index of refraction than that of the glass. In particular, 84 shows a ZMW structure, 85 indicates a ray of light being focused onto the ZMW by the integral lenses such as the embedded microlens.
100801 Figure 11 depicts one alignment strategy and optical system. The system comprises a photodetector 131, an optional lens 132 for collecting light, a ZMW 133 having a metal film 134 coupled to a substrate 135, and an objective lens 136 that is aligned with the incident light beam 137. Figure 13 depicts an exemplary alignment detection system and the associated components. The illustrative system 13A
comprises an optical confinement such as a zero-mode waveguide 111 having a metal film 113 coupled to a substrate 114. The zero-mode waveguide 111 typically contains signal generating molecules 112, and is optically linked to the associated components including an objective lens 115, a beam splitter/dichroic cube 117, optically a telen lens 120 (used in infinity corrected systems), and a photodetector 122 (e.g., a quadrant photodetector).
116 depicts rays propagating though system. 118 depicts the incident illumination rays. 119 depicts the return rays moving towards the detector 122.
Figure 13B depicts a front view of the quadrant photodiode. Shown in the center of the figure is a beam mis-aligned on the center of the quadrant detector. The four voltages generated by the four quadrants can be processed to determine the degree and direction of mis-alignment of the beam and thus the optical confinement such as ZMW
111.
100811 The subject arrays may comprise a single row or a plurality of rows of optical confinements on the surface of a substrate, where a plurality of lanes are present, for example, usually at least 2, more commonly more than 10, and more commonly more than 100. The subject array of optical confinements may align horizontally or diagonally long the x-axis or the y-axis of the substrate. The individual confinements can be arrayed in any format across or over the surface of the substrate, such as in rows and columns so as to form a grid, or to form a circular, elliptical, oval, conical, rectangular, triangular, or polyhedral pattern. To minimize the nearest-neighbor distance between adjacent optical confinements, a hexagonal array is preferred.
[0082] The array of optical confinements may be incorporated into a structure that provides for ease of analysis, high throughput, or other advantages, such as in a microtiter plate and the like. Such setup is also referred to herein as an "array of arrays." For example, the subject arrays can be incorporated into another array such as microtiter or multi-well plate wherein each micro well of the plate contains a subject array of optical confinements. Typically, such multi-well plates comprise multiple reaction vessels or wells, e.g., in a 48 well, 96 well, 384 well or 1536 well format. In such cases, the wells are typically disposed on 18 mm, 9 mm, 4.5 mm, or 2.25 mm centers, respectively.
[0083] An illustrative array of arrays is depicted in Figure 5 in which a subarray 71 is part of a super array 72.
Arrays can also be arranged in lattices. For example, Figure 4 depicts a top view of an illustrative regular disposition of ZMWs. In this configuration, there is a lattice defined by the parameters dl, d2, and the angle 53. In addition to a ZMW at each lattice point, there is a complex unit cellthat comprises a plurality of ZMWs in an arrangement that is defined by a list of angles and distances with one angle and one distance for each element of the unit cell. In particular, 52 represents the first lattice distance, 53 represents the lattice angle, 54 represents the second lattice distance, 55 represents the unit cell first distance, and 56 represents unit cell first angle. While this figure shows an array with a unit cell of two components, the unit cell can have any plurality of elements.
[0084] As described above, the subject arrays comprise a plurality of optical confinements. In some embodiments, the arrays have at least about 20 X 104 distinct optical confinements, preferably at least about 20 X
106 distinct confinements, and more preferably at least about 20 X 108 confinements. The density of the spots on the solid surface in certain embodiments is at least above 4 X104 confinements per mm2, and usually at least about 8 X104, at least about 1.2 x 105, or at least about 4 x 106 confinements per mm2, but does not exceed 4 X1012 confinements per mm2, and usually does not exceed about 4 X101 confinements per mm2. The overall size of the array generally ranges from a few nanometers to a few millimeters in thickness, and from a few millimeters to 50 centimeters in width or length. Preferred arrays have an overall size of about few hundred microns in thickness and may have any width or length depending on the number of optical confinements desired.
[0085] In one example as shown in Figure 1, the array of optical confinements, e.g. zero-mode waveguides, are arranged in a square format. The array comprises a representative zero-mode waveguide 21, separated from an adjacent waveguide by a distance "d" (22 represents the inter-zero mode waveguide spacing). In another example as shown in Figure 2, the array of optical confinements, e.g. zero-mode waveguides, are arranged in a non-square =

format. The array comprises a representative zero-mode waveguide 31, separated from an adjacent waveguide by a distance "d" (32 represents the inter-zero mode waveguide spacing). 33 shows the angle formed between any three adjacent ZMWs (e.g., 60 degrees). Figure 3 depicts a top view of another illustrative 2-dimentional array. The adjacent optical confinements are separated in one dimension by a distance of "dl" and in another dimension by a distance of "d2", with a unit vector angle 43.
100861 The spacing between the individual confinements can be adjusted to support the particular application in which the subject array is to be employed. For instance, if the intended application requires a dark-field illumination of the array without or with a low level of diffractive scattering of incident wavelength from the optical confinements, then the individual confinements are typically placed close to each other relative to the incident wavelength.
100871 Accordingly, in one aspect, the present invention provides an array of zero-mode waveguides comprising at least a first and at least a second zero-mode waveguide, wherein the first zero-mode waveguide is separated from the second zero-mode waveguide by a distance such that upon illumination with an incident wavelength, intensity of diffractive scattering observed from the first zero-mode waveguide at a given angle is less than that if the first zero-mode waveguide were illuminated with the same incident wavelength in the absence of the second zero-mode waveguide. Diffractive scattering can be reduced or significantly eliminated if an array comprises zero-mode waveguides spaced in a regular spaced lattice where the separation of zero-mode waveguides from their nearest neighbors is less than half the wavelength of the incident wavelength. In this regime, the structure behaves as a zero-order grating. Such gratings are incapable of scattering incident light despite having a large number of elements that by themselves would scatter very effectively. This arrangement is highly desirable for illumination approaches such as dark field illumination, where surface scattering would cause excitation radiation to be collected by the objective lens, thus increasing background noise. Useful wavelengths for illumination range from 250 nm up to 8 microns, meaning that an array of zero-mode waveguides with a spacing of less than 4000 nm would still be useful for application in this manner. A spacing of less than 2000 nm is more preferable, while a spacing of less than 1000 nm is even more preferable in this respect. Some configurations with spacing larger than one half of the wavelength can have the same advantage if the illumination is applied asymmetrically, or if the collection cone angle is configured to be less than 90 degrees. In addition to the benefit of reduced diffractive scattering, narrow spacing between the individual confinements decreases the illumination area and thus lowers the power demand.
[0088] Arrays having the optical confinements spaced far apart relative to the incident wavelength also have desirable properties. While the angle-dependent scattering raises the background signal that could be disadvantageous for certain applications, it provides a means particularly suited for characterizing the size and shape of the optical confinements. It also readily permits ensemble bulk measurements of molecule interactions, involving especially unlabelled molecules. Arrays suited for such applications generally contain individual confinements separated by more than one wavelength of the incident radiation, usually more than 1.5 times the incident wavelength, but usually does not exceed 150 times the incident wavelength.
Kits:
100891 The present invention also encompasses kits containing the optical confinement arrays of this invention.
Kits embodied by this invention include those that allow characterizing molecules and/or monitoring chemical reactions at a single-molecule level. Each kit usually comprises the devices and reagents which render such characterization and/or monitoring procedure possible. Depending on the intended use of the kit, the contents and packaging of the kit will differ. Where the kit is for DNA sequencing, the kit typically comprises: (a) an array of optical confinements, preferably zero-mode waveguides of the present invention, that permits resolution of individual molecules or the reaction of individual molecules, such as those that are present at a concentration higher than about 1 micromolar; (b) sequencing reagents typically including polymerases, aqueous buffers, salts, primers, and nucleotides or nucleotide analogs. Where desired a, 'control' nucleic acids of known sequence can be included to monitor the accuracy or progress of the reaction.
[00901 The reagents can be supplied in a solid form, immobilized form, and/or dissolved/suspended in a liquid buffer suitable for inventory storage, and later for exchange or addition into the reaction medium when the test is performed. Suitable individual packaging is normally provided. The kit can optionally provide additional components that are useful in the procedure. These optional components include, but are not limited to, buffers, capture reagents, developing reagents, labels, reacting surfabes, control samples, instructions, and interpretive information. Diagnostic or prognostic procedures using the kits of this invention can be performed by clinical laboratories, experimental laboratories, practitioners, or private individuals.
Preparation of the Optical Confinements:
[00911 The array of the present invention can be manufactured using nanofabrication techniques provided by the present invention, as well as those known in the fields of Integrated Circuit (IC) and Micro-Electro-Mechanical System (MEMS). The fabrication process typically proceeds with selecting an array substrate, followed by using appropriate IC processing methods and/or MEMS micromachining techniques to construct and integrate the optical confinement and other associated components.

Array Substrate:
[0092] In some embodiments, the array of optical confinements is present on a rigid substrate. In other embodiments concerning, e.g., porous films with reflective index media, flexible materials can be employed. In general, a rigid support does not readily bend. Examples of solid materials which are not rigid supports with respect to the present invention include membranes, flexible metal or plastic films, and the like. As such, the rigid substrates of the subject arrays are sufficient to provide physical support and structure to optical confinements present thereon or therein under the assay conditions in which the array is employed, particularly under high throughput handling conditions.
[0093] The substrates upon which the subject patterns of arrays are disposed, may take a variety of configurations ranging from simple to complex, depending on the intended use of the array.
Thus, the substrate could have an overall slide or plate configuration, such as a rectangular or disc configuration, where an overall rectangular configuration, as found in standard microtiter plates and microscope slides, is preferred. Generally, the thickness of the rigid substrates will be at least about 0.01 mm and may be as great as 1 cm or more, but will usually not exceed about 5 cm. Both the length and the width of rigid substrate will vary depending on the size of the array of optical confinements that are to be fabricated thereon or therein.
100941 The substrates of the subject arrays may be fabricated from a variety of materials. The materials from which the substrate is fabricated is preferably transparent to visible and/or UV light. Suitable materials include glass, semiconductors (e.g., silicate, silicon, silicates, silicon nitride, silicon dioxide, quartz, fused silica, and gallium arsenide), plastics, and other organic polymeric materials. In preferred aspects, silica based substrates like glass, quartz and fused silica are used as the underlying transparent substrate material.
[0095] The substrate of the subject arrays comprise at least one surface on which a pattern of optical confinements is present, where the surface may be smooth or substantially planar, or have irregularities, such as depressions or elevations. The surface may be modified with one or more different layers of compounds that serve to modulate the properties of the surface in a desirable manner. Modification layers of interest include: inorganic and organic layers such as metals, metal oxides, polymers, small organic molecules, functional moieties such as avidin/biotin and the like. The choice of methods for applying the coating materials will depend on the type of coating materials that is used. In general, coating is carried out by directly applying the materials to the zero-mode waveguide followed by washing the excessive unbound coating material from the surface. Alternatively or additionally, coating materials may be deposited using other conventional techniques, such as chemical vapor deposition (CVD), sputtering, spin coating, in situ synthesis, and the like. Certain coating materials can be cross-linked to the surface via heating, radiation, and/or by chemical reactions. In preferred aspects, suitable coating materials are coupled to substrate surfaces either covalently or through ionic or hydrophobic/hydrophilic interactions. In the case of silica based substrates, for example, silane chemistries are particularly suited for covalently attaching coating materials to surfaces, e.g., coupling groups, specific binding moieties, and the like. Such chemistries are well known to those of ordinary skill in the art and can be practiced without undue experimentation.
Fabrication Process:
[0096] Fabrication of the subject array substrates can be performed according to the methods described as follows or other standard techniques of IC-processing and/or MEMS micromachining. The standard techniques known in the art include:but are not limited to electron-beam lithography, photolithography, chemical vapor or physical vapor deposition, dry or wet etching, ion implantation, plasma etching, bonding, and electroplating. Additional fabrication processes are detailed in the U.S. Patent Application Publication No.
2003/0174992.
[00971 In a preferred embodiment, the present invention provides a negative tone fabrication process, which provides for the creation of optical confinements having more uniform and consistent dimensions than conventional positive tone fabrication processes that can yield optical confinements of varying dimensions. A comparison of the two fabrication processes is shown in Table 1 below.
Table 1 - Positive and Negative Tone Process Steps in Fabrication of Zero-Mode Waveguides Step # Positive Tone Process Negative Tone Process Clean fused silica substrates in heated solution of hydrogen Same peroxide and ammonium hydroxide.
2 Cascade rinse substrates in deionized water. Same 3 Clean substrates in oxygen plasma cleaner. Same Coat substrates with metal film by either thermal Spin-coat substrates with electron-evaporation or sputtering, beam resist.
Spin-coat substrates with electron-beam resist over the Bake casting solvent out of film.

metal layer.
Bake casting solvent out of film. Expose resist with electron beam lithography.
7 Expose resist with electron beam lithography. Develop resist in chemical bath to reveal array of small pillars with large empty gaps in resist.
Develop resist in chemical bath to reveal holes. Rinse developer away and dry chips.
Rinse developer away and dry chips. Coat chips with metal film by 9 either thermal evaporation or sputtering.
Use reactive-ion etching to transfer resist pattern into metal Dissolving underlying negative film. resist using Microposit 1165 Stripper.
11 Strip resist using oxygen plasma. Same [0098] In a negative tone process, a negative resist is applied to the substrate. A resist is negative if it is rendered insoluble by application of some agent, wherein the case of photoresists or e-beam resists, the agent is optical energy or electron beam energy, respectively. Alternatively, a positive tone resist can be used with a negative pattern. A
5 negative tone pattern is characterized by the application of the agent in all areas except the location of the optical confinement, e.g., zero-mode waveguide, contrasted with a positive tone image in which the agent is confined only to the optical confinement area. In either case, after development of the resist, resist remains only in the areas where the optical confmement is intended to lie. It is useful in many cases to use means to achieve an undercut sidewall profile of these remaining resist features. Many techniques exist in the art to obtain undercut sidewalls, for example, 10 in electron beam lithography. For instance, when using negative tone resists, one method is to apply to layers of electron beam resist to the surface sequentially, the upper film having a higher sensitivity to the energy delivered to it by the electron beam. Because the beam has a tendency to spread, a larger area of the upper film will be rendered insoluble than in the lower layer, resulting in an overhang beneath the upper layer as desired.
100991 After development and appropriate cleaning procedures known in the art such as a plasma cleaning procedure, the metal film comprising the optical confinement can be applied by one of several methods, including metal evaporation, molecular beam epitaxy and others. In the case that the resist profile is undercut as discussed above, the metal that is deposited in the regions still occupied by the resist will rest on top of the resist rather than resting on the device surface. The resist layer is subsequently removed by any of several techniques including solvent dissolution either with or without ultrasonication or other mechanical agitation, reactive plasma etching, vaporization or others. The metal which rested on the resist features is removed as the resist is removed ("lifted off'), while the resist resting directly on the substrate remains to form the walls of the optical confinement.
[001001 The advantage of this process is that the size of the optical confinement is determined by the size of the resist feature, and does not rely on the fidelity of reactive ion etch pattern transfer mechanisms, which can be highly variable for metal films, especially aluminum a desirable metal for these devices. The positive tone process is subject to the inherent variation in resist feature sizes plus the variation due to pattern transfer, while the negative tone process is subject to the first variability but not the second. Metal thin film techniques suffer from much less lateral variation, and so the overall accuracy is better. This method also does not rely on the availability of a suitable etch for the metal in question, allowing the application of the process to a much wider selection of metals than the positive tone process.
1001011 Figure 6 is a schematic presentation of an illustrative negative tone process to make zero-mode waveguides. In this process, the substrate 11 is first coated with a layer of negative resist 12. Optionally, the substrate can be coated with a second resist layer 13. Exposure of the resist to the same pattern electron beam lithography tool used in the positive tone process, generates the opposite pattern as previously observed, namely one of a periodic array of small pillars of remaining resist, and empty gaps between the pillars 15. The final zero-mode waveguide structures are created by coating this pattern with a thin metal layer such as an aluminum layer 17, and then dissolving the underlying negative resist pillars 18. Because this process is not dependent on the thickness of the alumina layer or the crystal structure or morphology of the metal film, it produces a far more consistent configuration, and provides much finer control over the critical feature size.
Figure 8 depicts a scanning electron micrographs of ZMW structures fabricated by positive tone resist (left panels) or negative tone resist (right panels).
The grain structure of the polycrystalline film is visible in the image as flecks, and the ZMWs as dark round structures.
[001021 A variant negative tone process is termed nanocasting. The steps of nanocasting are similar except that the use of bi-layer resist is avoided. The process first involves depositing on the surface of a substrate (in this case a single-layer resist would be used). The electron beam exposure and development follow, leaving a cylindrical feature for each dot in the exposure pattern. For this process, it is desirable to allow the metal deposition technique to apply material not just on the top of the resist structure but also on the sidewalls of the resist feature. This process is inherently three dimensional, in that a negative replica of the exterior surface of the three-dimensional resist feature is reproduced in the interior surface of the metal films that forms the optical confinement walls. In this case, the undercut resist profile and the various methods used to produce this are not necessary, as in the negative tone process, they are used specifically to prevent contact of the deposited film with the sides of the resist feature. In the nanocasting approach, the deposited film faithfully reproduces the exterior surface of the resist feature, so an undercut figure would only be used if a non-cylindrical confinement is desired.
[00103] In practicing nanocasting, caution is typically employed to removed the metal from above the nanocasting "master" (the resist feature), as the resist feature can in some instances be entirely buried and unavailable for removal. This, however, can be remedied in a number of ways.
[00104] Where the deposition technique has a high degree of anisotropy in the deposition (such as metal evaporation), the sidewalls will be very thin near the top of the resist feature, which in some instances can be a cylindrical pillar. This weak point can be subject to direct mechanical disruption allowing the removal of the metal above the resist feature and hence the ZMW location. An isotropic etch, either solution phase or plasma can be used to further thin the film until this weak point separates, achieving the same effect. If the metal deposition step has a low degree of anisotropy (such as sputtering or electroplating), then the resist material can be exposed through chemical mechanical polishing, or ion milling.
[00105] Simultaneous with or subsequent to the removal of the metal cap over the resist feature, the resist material is then removed by solvent dissolution, or reactive ion etching. This completes the fabrication steps, provided the appropriate pattern is applied and the other parameters are correctly chosen.
Uses of the Subject Optical Confinements and Other Devices:
[00106] The subject devices including optical confinements and associated optical systems provide a effective means for analyzing molecules and monitoring chemical reactions in real time.
The subject device and organism, determining comprehensive mutation sets for individual clones and profiling in various diseases or disease stages. Other applications include measuring enzyme kinetics, and identifying specific interactions between target molecules and candidate modulators of the target molecule. Further applications involve profiling cell receptor diversity, identifying known and new pathogens, exploring diversity towards agricultural, environmental and therapeutic goals.

[00107] In certain embodiments, the subject devices and methods allow high-throughput single-molecule analysis.
Single-molecule analysis provides several compelling advantages over conventional approaches to studying biological events. First, the analysis provides information on individual molecules whose properties are hidden in the statistically averaged information that is recorded by ordinary ensemble measurement techniques. In addition, because the analysis can be multiplexed, it is conducive to high-throughput implementation, requires smaller amounts of reagent(s), and takes advantage of the high bandwidth of optical systems such as modern avalanche photodiodes for extremely rapid data collection. Moreover, because single-molecule counting automatically generates a degree of immunity to illumination and light collection fluctuations, single-molecule analysis can provide greater accuracy in measuring quantities of material than bulk fluorescence or light-scattering techniques.
As such, single-molecule analysis greatly improves the efficiency and accuracy in genotying, gene expression profiling, DNA sequencing, nucleotide polymorphism detection, pathogen detection, protein expression profiling, and drug screening.
Single-Molecule Sequencing:
[00108] The subject devices, including various forms of optical confinements and the associated optical systems, are particularly suited for multiplexed single-molecule sequencing.
Accordingly, the present invention provides a method of simultaneously sequencing a plurality of target nucleic acids. The method generally involves (a) providing an array of optical confinements of the present invention; (b) mixing in the confinements a plurality of target nucleic acid molecules, primers complementary to the target nucleic acid molecules, polymerization enzymes, and more than one type of nucleotides or nucleotide analogs to be incorporated into a plurality of nascent nucleotide strands each being complimentary to a respective target nucleus and molecules;
(c) subjecting the mixture to a polymerization reaction under conditions suitable for formation of the nascent nucleotide strands by template-directed polymerization; (d) illuminating the waveguides with an incident light beam; and (e) identifying the nucleotides or the nucleotide analogs incorporated into each nascent nucleotide strand.
[00109] The subject sequencing methods can be used to determine the nucelic acid of any nucleic acid molecule, including double-stranded or single-stranded, linear or circular nucleic acids (e.g., circular DNA), single stranded DNA hairpins, DNA/RNA hybrids, RNA with a recognition site for binding of the polymerase, or RNA hairpins.
The methods of the present invention are suitable for sequencing complex nucleic acid structures, such as 5' or 3' non-translation sequences, tandem repeats, exons or introns, chromosomal segments, whole chromosomes or -genomes.

1001101 In one aspect, the temporal order of base additions during the polymerization reaction is identified on a single molecule of nucleic acid. Such identifying step takes place while the template-directed extension of primer or polymerization is taking place within the optical confinement. In a preferred embodiment, single-molecule sequencing is performed in a homogenous assay that does not require transfer, separation, or washing away any reactant or by-product (e.g. fluorophore cleaved from a nucleotide) after each base addition event. In certain aspects of the homogenous assay, single-molecule sequencing is performed without adding reactants to the mixture prior to reading the next base sequence. In this assay, stepwise addition of nucleotides or removal of by-products after each base addition event is not necessary, as diffusion of reactants from a large volume of reagents above the confinement will not interfere with the detection of incorporation. Sequence information is generated continuously as the polymerase continually incorporates the appropriate nucleotides or nucleotide analogs into the nascent DNA strand.
For a detailed discussion of such single molecule sequencing, see, e.g., Published U.S. Patent Application No.
2003/0044781, and M.J. Levene, J. Korlach, S.W. Turner, M. Foquet, H.G. Craighead, W.W. Webb, SCIENCE 299:682-686, January 2003 Zero-Mode Waveguides for Single-Molecule Analysis at High Concentrations. There is no loss of synchronization because single molecules are observed separately. This method also allows the use of target nucleic acid molecules taken directly from a biological sample, minimizing the need for cloning, subcloning, or amplification of the target nucleic acids before sequencing can take place.
1001111 In a preferred embodiment, a polymerase enzyme is provided anchored within the effective observation volume within an optical confinement. Template dependent synthesis of a complementary strand is then carried out while observing the volume, and using labeled nucleotide analogs that are capable of being sequentially incorporated into the growing strand without interruption, e.g., for deprotection, etc. In preferred aspects, nucleotide analogs bearing a label on a non-incorporated phosphate group or derivative, e.g., the beta, gamma, delta, etc.
phosphate of a nucleotide polyphosphate which is cleaved from the analog during incorporation, are used in such methods. Such nucleotide analogs provide an advantage of being sequentially incorporated into the growing nucleic acid strand, and having their labeling groups removed in the incorporation process so as to not provide increasing signal noise during synthesis that would result if such labels remained associated with the synthesized strand. In addition, because the incorporation event provides for prolonged presence of the labeled analogs within the observation volume (as compared to random diffusion of non-incorporated analogs into the observation volume), the signal associated within incorporation is readily identifiable. In particularly preferred aspects, for single molecule nucleic acid sequencing applications, a template nucleic acid is used that provides for the redundant or iterative reading/synthesis of tandem repeats of a particular sequence segment of interest. In particular, the systems of the invention typically provide for redundancy in numerous ways, to correct for any errors that may arise in template dependant synthesis by the polymerase enzyme. For example, because the methods of the invention focus on single molecules, redundant processes are employed to assure that mis-incorporation events by a polymerase are corrected for in data analysis.
[00112] In a first aspect, such redundancy is supplied by utilizing arrays of multiple different confinements that are being applied to a given sequence of interest, e.g., in a single well of a multi-well plate. In addition to this redundancy, the invention also provides for the iterative sequencing of a given sequence segment (or a copy thereof) multiple times within a single confinement. In a first preferred aspect, such iterative sequencing may be accomplished by providing the sequence segment of interest in a circular template format, so that the polymerase processes around the circular template (allowing the elucidation of the sequence of such template) multiple times.
Methods of circularization of nucleic acid segments are known to those of ordinary skill in the art, and are readily applied to template sequences in accordance with the invention.
[00113] In another aspect, a similar result is accomplished by using a template-dependant circular template bearing the sequence segment of interest. In particular, such synthesis product will typically include, in a single linear strand, multiple copies of the circular template, again, providing for iterative sequencing of the sequence segment of interest. Further, redundancy is additionally accomplished by circularizing this linear, multi-copy template and iteratively sequencing multiple copies, multiple times.
[00114] In another aspect, a similar result is obtained by performing concatmerization of amplicons generated in a single-molecule amplification strategy, several of which are known to those skilled in the art. These strategies can employ dilution to the single molecule level, or isolation of molecules in small micelles in a two-phase emulsion during amplification. The concatmerized strand is then sequenced as a single template, and redundant information is generated from a single molecule in this fashion.
[00115] In yet another aspect, a similar result is obtained by using a long double stranded template with nicks and/or gaps at multiple locations along it. The molecule can then be caused to initiate single molecule sequencing at several locations along the strand, each location comprising a confinement that independently sequences the strand.
Because the several confinements are acting on the same strand, the result is that the same template is sequenced several times providing redundant information from a single molecule.

Exemplary Experimental Setup:
[00116] In practicing a sequencing method of the present invention, a reaction mixture comprising the target nucleic acid(s) of interest, primers complementary to the target nucleic acids, polymerization enzymes, and more than one type of nucleotides or nucleotide analogs, is applied to an array of optical confinements. Preferably, each optical confinement receives only one target nucleic acid molecule that is to be sequenced. This can be achieved by diluting a minute amount of target nucleic acids in a large volume of solution containing the rest of the reactants required for the sequencing process. Alternatively, a non-cylindrical waveguide whose the opening is narrower in lateral dimension than the base, can be used to restrict the entry of multiple target nucleic acids.
Immobilization of the target nucleic acid or the polymerase to an optical confinement:
[00117] The target nucleic acid can be immobilized to the inner surface of the optical confinement by a number of ways. For example, the target nucleic acid can be immobilized onto an optical confinement by attaching (1) a primer or (2) a single-stranded target nucleic acid or (3) double-stranded or partially double-stranded target nucleic acid molecule. Thereafter, either (1) the target nucleic acid molecule is hybridized to the attached oligonucleotide primer, (2) an oligonucleotide primer is hybridized to the immobilized target nucleic acid molecule to form a primed target nucleic acid molecule complex, or (3) a recognition site for the polymerase is created on the double-stranded or partially double-stranded target nucleic acid (e.g., through interaction with accessory proteins, such as a primase).
A nucleic acid polymerizing enzyme on the primed target nucleic acid molecule complex is provided in a position suitable to move along the target nucleic acid molecule and extend the oligonucleotide primer at the site of polymerization.
[00118] In preferred aspects, as described previously, the polymerization enzyme is first attached to a surface of the subject optical confinement within the effective observation volume of the confinement, and in a position suitable for the target nucleic acid molecule complex to move relative to the polymerization enzyme.
[00119] One skilled in the art will appreciate that there are many ways of immobilizing nucleic acids and enzymes onto an optical confinement, whether covalently or noncovalently, via a linker moiety, or tethering them to an immobilized moiety. These methods are well known in the field of solid phase synthesis and micro-arrays (Beier et al., Nucleic Acids Res. 27:1970-1-977 (1999). Non-limiting exemplary binding moieties for attaching either nucleic acids or polymerases to a solid support include streptavidin or avidin/biotin linkages, carbamate linkages, ester linkages, amide, thiolester, (N)-functionalized thiourea, functionalized maleimide, amino, disulfide, amide, hydrazone linkages, and among others. Antibodies that specifically bind to the target nucleic acids or polymerases can also be employed as the binding moieties. In addition, a silyl moiety can be attached to a nucleic acid directly to a substrate such as glass using methods known in the art.
1001201 Where desired, the polymerases may be modified to contain one or more epitopes such as Myc, HA
(derived from influenza virus hemagglutinin), poly-histadines, and/or FLAG, for which specific antibodies are available commercially. In addition, the polymerases can be modified to contain heterologous domains such as glutathione S-transferase (GST), maltose-binding protein (MB?), specific binding peptide regions (see e.g., U.S.
Patent Nos. 5,723,584, 5,874,239 and 5,932,433), or the Fc portion of an immunoglobulin. The respective binding agents for these domains, namely glutathione, maltose, and antibodies directed to the Fc portion of an immunoglobulinxe available, and can be used to coat the surface of an optical confinement of the present invention.
1001211 The binding moieties or agents of either the polymerases or nucleic acids they immobilize can be applied to the support by conventional chemical techniques which are well known in the art. In general, these procedures can involve standard chemical surface modifications of a support, incubation of the support at different temperature levels in different media comprising the binding moieties or agents, and possible subsequent steps of washing and cleaning.
Reaction Mixture: Labeled Nucleotides, Polym erases, and Primers:
1001221 The various types of nucleotides utilized in accordance with the single-molecule sequencing method are conjugated with detectable labels so that a photon detector can detect and distinguish their presence within the subject optical confinements. Preferred labels are luminescent labels, and especially fluorescent or chromogenic labels.
1001231 A variety of functional groups used as detectable labels in nucleotides has been developed in the art. Table 1 lists numerous examples of such functional groups. Additional examples are described in U.S. Patent No.
6,399,335, published U.S. Patent Application No. 2003/0124576, and The Handbook ¨ 'A Guide to Fluorescent Probes and Labeling Technologies, Tenth Edition' (2005) (available from Invitrogen, Inc.,/Molecular Probes), Table 1 Exemplary detectable label functional groups 4-aminophenol 6-aminonaphthol 4-nitrophenol 6-nitronaphthol 4-methylphenol 6-chloronaphthol 4-methoxyphenol 6-bromonaphthol 4-chlorophenol 6-iodonaphthol 4-bromophenol 4,4'-dihydroxybiphenyl 4-iodophenol 8-hydroxyquinoline 4-nitronaphthol 3-hydroxypyridine 4-aminonaphthol umbelliferone 4-methylnaphthol Resorufin 4-methoxynaphthol 8-hydroxypyrene 4-chloronaphthol 9-hydroxyanthracene 4-bromonaphthol 6-nitro9-hydroxyanthracene 4-iodonaphthol 3-hydroxyflavone 6-methylnaphthol fluorescein 6-methoxynaphthol 3-hydroxybenzoflavone 1001241 Using these or other suitable functional groups known in the art, a vast diversity of fluorophores suitable for the present sequencing method can been generated. They include but are not limited to 4-acetamido-4'-isothiocyanatostilbene-2,2'disulfonic acid, acridine and derivatives such as acridine and acridine isothiocyanate, 5-(2'-aminoethyl)aminonaphthalene-1-sulfonic acid (EDANS), 4-amino-N[3-vinylsulfonyl)phenylinaphthalimide-3,5 disulfonate (Lucifer Yellow VS), N-(4-anilino-l-naphthyl)maleimide, anthranilamide, Brilliant Yellow, coumarin and derivatives such as coumarin, 7-amino-4-methylcoumarin (AMC, Coumarin 120), 7-amino-4-trifluoromethylcouluarin (Coumaran 151); cyanosine; 4',6-diaminidino-2-phenylindole (DAPI); 5, 5"-dibromopyrogallol-sulfonephthalein (Bromopyrogallol Red); 7-diethylamino-3-(4'-isothiocyanatopheny1)-4-methylcoumarin; diethylenetriamine pentaacetate; 4,4'-diisothiocyanatodihydro-stilbene-2,2'-disulfonc acid; 4,4'-diisothiocyanatostilbene-2,2'-disulfonic acid; 54dimethylamino]naphthalene-1-sulfonyl chloride (DNS, dansyl chloride); 4-(4'-dimethylaminophenylazo)benzoic acid (DABCYL); 4-dimethylaminophenylazopheny1-4'-isothiocyanate (DABITC); eosin and derivatives such as eosin and eosin isothiocyanate; erythrosin and derivatives such as erythrosin B and erythrosin isothiocyanate; ethidium; fluorescein and derivatives such as 5-carboxyfluorescein (FAM), 5-(4,6-dichlorotriazin-2-yl)aminofluorescein (DTAF), 2'7'-dimethoxy-4'5'-dichloro-6-PCT/US2005/0330.52 carboxyfluorescein (JOE), fluorescein, fluorescein isothiocyanate (FITC), and QFITC (XR1TC); fluorescamine;
IR144; IR1446; Malachite Green isothiocyanate; 4-methylumbelliferone; ortho cresolphthalein; nitrotyrosine;
pararosaniline; Phenol Red; B-phycoerythrin; o-phthaldialdehyde; pyrene and derivatives such as pyrene, pyrene butyrate and succinirnidyl 1-pyrene butyrate; Reactive Red 4 (Cibacron ®
Brilliant Red 3B-A); rhodamine and derivatives such as 6-carboxy-X-rhodamine (ROX), 6-carboxyrhodamine (R6G), lissamine rhodamine B sulfonyl chloride, rhodamine (Rhod), rhodamine B, rhodamine 123, rhodamine X
isothiocyanate, sulforhodamine B, sulforhodamine 101 and sulfonyl chloride derivative of sulforhodamine 101 (Texas Red); N,N,N,N'-tetramethy1-6-carboxyrhodamine (TAMRA); tetramethyl rhodamine; tetramethyl rhodamine isothiocyanate (TRITC); riboflavin;
rosolic acid and terbium chelate derivatives. Additional fluorophores applicable for the subject sequencing methods are disclosed in U.S. patent no. 5,866,366 and WO 01/16375.
(001251 The labels can be attached to the phosphate backbone, on the base, on the ribose unit, or a combination thereof. Preferred labels are those that do not substantially impede the continuous addition of nucleotides in a sequencing reaction. Such labels include those linked to the alpha phosphate, the beta phosphate, the terminal phosphate, or the delta or more distal phosphates in tetra, penta or hexa phosphate nucleotides, or the base unit of a nucleotide.
[001261 Nucleotides comprising labeled terminal phosphates (e.g., the gamma phosphate as in dNTP), are particularly preferred because no additional means is required to remove the label in the sequencing procedure.
During the process of nucleic acid polymerization, the bond cleavage in the nucleotide occurs between the alpha and the beta phosphate, causing the beta and terminal phosphate (e.g., the gamma phosphate as in dNTP) to be released from the site of polymerization. As such, the label attached to the terminal phosphate is separated from the nascent strand once the nucleotide is being incorporated. In general, terminal-phosphate-linked nucleotides may comprise three or more phosphates, typically about three to about six phosphates, preferably about three to about five phosphates. Table 1 lists numerous examples of nucleotides with labeled terminal phosphates. Many other terminal-phosphate-linked nucleotides have been developed and are detailed in U.S. patent application number 2003/024576.
Table 2 Adenosine-5'-()-4-nitrophenyl)triphosphate Guanosine-5'47-4-nitrophenyptriphosphate Cytosine-5'-(y-4-nitrophenyptriphosphate Thymidine-5'47-4-nitropheny1)triphosphate Uracil-5'-(y-4-nitrophenyptriphosphate 3'-azido-31-deoxythymidine-5'-(7-4-nitropheny1)triphosphate 3'-azido-2',3'-dideoxythymidine-5'-(7-4-nitrophenyl)triphosphate 2',3'-didehydro-2',3'-dideoxythymidine-5'-(y-4-nitrophenyl)triphosphate Adenosine-5'-(7-4-aminophenyl)triphosphate Adenosine-5'47-4-methylphenyl)triphosphate Adenosine-5'47-4-methoxyphenyptriphosphate Adenosine-5'47-4-chlorophenyptriphosphate Adenosine-5'-(y-4-bromophenyl)triphosphate Adenosine-5'47-4-iodophenyl)triphosphate Adenosine-5'47-4-nitronaphthyl)triphosphate Adenosine-5'-(y-4-aminonaphthyl)triphosphate Adenosine-5'47-4-methylnaphthyl)triphosphate Adenosine-5'-(y-4-methoxynaphthyl)triphosphate Adenosine-5'47-4-chloronaphthyptriphosphate Adenosine-5'-('y-4-bromonaphthyl)triphosphate Adenosine-51-(7-4-iodonaphthyptriphosphate Adenosine-5'47-6-methylnaphthyptriphosphate Adenosine-5'-ey-6-methoxynaphthy1)triphosphate Adenosine-5'47-6-aminonaphthyptriphosphate Adenosine-5'-(y-6-nitronaphthyl triphosphate Adenosine-5'-('y-6-chloronaphthyptriphosphate Adenosine-5'-(y-6-bromonaphthyl)triphosphate Adenosine-5'47-6-iodonaphthyl)triphosphate Adenosine-5'-(7-4'-hydroxybiphenyptriphosphate Adenosine-51-(7-8-quinolyptriphosphate Adenosine-5'-(7-3-pyridyptriphosphate Adenosine-5'-(y-umbelliferone)triphosphate Adenosine-5'(y-resorufin)triphosphate Adenosine-5'-(y-pyrene)triphosphate Adenosine-5'-(y-anthracene)triphosphate Adenosine-5'-(y-6-nitroanthracene)triphosphate Adenosine-5'-('y-flavonyl)triphosphate Adenosine-5'(7-fluorescein)triphosphate Adenosine-51-(7-benzoflavone)triphosphate Adenosine-5'-(y-(4-nitropheny1)-y-(4-arninophenyptriphosphate Adenosine-5'-ey-(4-nitropheny1)-y-(4-nitronaphthyptriphosphate 1001271 Nucleotides comprising modified phosphate backbones can also be used.
For example, the modified component can be a phosphordiamidate, methylphosphonate, alkyl phosphotriester, formacetal, phosphorodithioate, phosphothioate, phosphoramidothioate, phosphoramidate, or an analog thereof.
[00128] In some embodiments, the nucleotides or nucleotide analogs used in the present invention are reversible extension terminators comprising reversible blocking groups. In some embodiments, the blocking group on a reversible extension terminator is linked to a detectable label. In other embodiments, the blocking group and the detectable label are located on different positions of a nucleotide. In yet other embodiments, the blocking group is also a label.
1001291 An illustrative reversible extension terminator comprises a labeled ribose unit at the 3' end. Each label on the ribose unit, typically acts as a reversible blocking group that must be removed before the next nucleotide addition event can take place during a polymerization reaction. Preferred 3'-ribose labels comprise photo-removable functional groups that can be deprotected upon exposure to a light beam at a suitable wavelength.
[00130] In another example, the reversible blocking group is located at the 2' or the 4' position of the ribose unit of a nucleotide. In yet another embodiment, the reversible blocking group is linked to or conjugated to the base (adenine, thymine, cytosine, guanine, or uracil) a nucleotide. Non-limiting examples of reversible blocking groups, and especially photocleavable blocking groups include but are not limited to those molecules depicted in Figures 14 and 15 and those described in the co-pending application serial no 60/649,009.
[00131] The wavelength used to cleave the photocleavable blocking groups will depend on the choice of the blocking group. The wavelength may range from about 320 mu to about 800 nm. In some embodiment, the wavelength for cleaving the blocking group is about the same as the wavelength used to detect the label. In other embodiments, the wavelength for cleaving the blocking group is different from the wavelength used to detect the label.
1001321 In some embodiments, it is advantageous to use a mixture of labeled nucleotides that is substantially free of unlabeled nucleotides. Such composition and the uses thereof for sequencing are detailed in co-pending application serial number 60/651,846. Briefly, the composition is prepared by treating a mixture comprising labeled and unlabeled nucleotides or nucleotide analogs with an agent that specifically modifies unlabeled or incorrectly labeled nucleotides or nucleotide analogs to reduce their ability to be used in a hybridization or sequencing assay. Preferably, the agent used specifically modifies unlabeled or incorrectly labeled nucleotides analogs to render them incapable of being used in a hybridization or sequencing assay. For example, the nucleotides can be modified so that they no longer contain structures generally needed for the Watson Crick base pairing in a hybridization or template-directed sequencing assay. In some embodiments, for example, base units of the nucleotides are modified. In some embodiments, phosphate groups, preferably terminal phosphate groups, of the nucleotides or nucleotide analogs are modified to yield molecules that are incorporated to a lesser extent into a nascent nucleic acid strand during a template-directed polymerization reaction. In more preferred embodiments, the terminal phosphate groups of a nucleotide or nucleotide analogs are modified to yield molecules that cannot or that substantially cannot be incorporated into a nascent nucleic acid strand during a template-directed polymerization reaction.
[00133] The agents can comprise one or more enzymes. A variety of enzymes known in the art are suitable for modifying the nucleotides or nucleotide analogs, e.g. by cleaving or altering the configuration of the sugar, base, or phosphates, so as to disrupt the specific Watson Crick base pairing. Exemplary agents include but are not limited to guanine or adenine P-ribosyl transferase, purine nucleoside phosphorylase, AMP
nuleosidase, nucleoside deoxyribosyl transferase for purines, and orotate P-ribosyl transferase, thymidine phosphorylase, thymidine or uridine nucleosidase, uridine phosphorylase, pyrimidine nucleoside phosphorylase nucleoside deoxyribosyl transferase.
[00134] Enzymes applicable for modifying the terminal phosphate groups of nucleotides or nucleotide analogs include a wide array of phosphatases. An example of such enzyme is Shrimp Alkaline Phosphatase (SAP) that can remove the gamma and beta phosphates from a deoxynucleoside triphosphate (dNTP). The enzyme can convert specifically unlabeled dNTP into a nucleoside monophosphate dNMP which is generally incapable of being utilized by a polymerase enzyme in a template-directed sequencing reaction. It has been shown, that this phosphatase selectively modify nucleotides that are not labeled, e.g. at the terminal phosphate. Therefore, in a mixture of terminal phosphate-labeled and unlabeled nucleotides, the SAP will preferentially act on unlabeled nucleotides, leaving a larger proportion of labeled nucleotides available for incorporation in a sequencing reaction.
[00135] Other suitable phosphatases that can be used include but are not limited to calf intestinal alkaline phosphatases, and/or phosphatases of other mammals, crustaceans, and other animals. Examples of phosphatases that may be useful practicing the present invention can be found in US
20040203097, US 20040157306, US
20040132155; and US 20040110180.
[00136] Any other naturally occurring or synthetic phosphatases or phosphatases made by recombinant DNA
technology can also be used so long as they specifically or preferentially convert unlabeled nucleotides or analogs (as compared to labeled nucleotides), to molecules that are substantially incapable of being utilized by a polymerization enzyme. Directed molecular evolution can also be used to enhance and extend the activity of related enzymes to yield the desired property described above. A wide variety of mutagenesis techniques, both in silicon and in situ, are available in the art. An example of a mutagenesis or screening assay for generating such enzymes can involve a first test for abrogation of polymerization in the system with unlabeled nucleotides, and a second screen checking for the retention of polymerization activity in the presence of labeled nucleotides. Both of these screens can be performed in the context of a highly multiplexed parallel assay. Enzymes showing some beneficial specificity can be retained, mutated by some method, and then re-screened.
Methods such as these have been shown to produce many orders of magnitude improvement in specificity and performance.
[00137] Enzymes capable of selectively or preferentially modifying a subset of unlabeled nucleotides can also be employed. For example, creatine kinase enzyme is specific for the removal of a phosphate from adenoside triphosphate, and will not act on other bases. Other enzymes that selectively or preferentially act on one or more types of unlabeled nucleotides can also be used.
1001381 The nucleotide modifying enzymes described above can be used to pre-treat the nucleotides or nucleotide analogs, or can be used in the hybridization and/or sequencing reaction mixture, e.g., along with other hybridization or sequencing reagents.
[00139] The reaction conditions under which the modification of the nucleotides takes place will vary depending on the choice of the modifying enzymes. In one aspect, the conditions may be set within the following parameters: pH
is between 4.0 and 12.0, more preferably between pH 6.0 and 10.0, more preferably between 7.0 and 9.0, more preferably less than 8, more preferably between 7 and 8, and most preferably pH 7.5 and 8.5, preferably controlled by a buffer. The buffer can be Tris-based preferably at pH 7.5 to pH 8.5.
Other buffers may be used such as, but not limited to: organic buffers such as MOPS, HEPES, TRICINE, etc., or inorganic buffers such as phosphate or acetate. Buffers or other agents may be added to control the pH of the solution thereby increasing the stability of the enzymes. Where desired, reducing agent such as but not limited to dithiotreitol (DTT) or 2-mercaptoethanol may be added to limit enzyme oxidation that might adversely affect stability of the enzymes. The choice of specific reaction conditions including various buffers and pH conditions is within the skill of practitioners in the field, and hence is not further detailed herein.
[00140] Upon completion of the pre-treatment, the enzymes can be heat-inactivated by raising the reaction temperature to at least about 65 C, preferably between about 65 C to about 80 C. Alternatively, the enzymes can be depleted from the reaction mixture by, e.g., centrifugation through a filter (e.g., Millipore) that has a molecular weight cutoff smaller than the size of the enzyme.
[00141] After the treatment, the mixture generally comprises less than about 30%, preferably less than about 20%, more preferably less than about 10%, more preferably less than about 5%, more preferably less than about 1%, more preferably less than about 0.5%, or more preferably less than about 0.1%, and even more preferably less than 0.01%
of unlabeled nucleotides or unlabeled nucleotide analogs. This enriched mixture of labeled nucleotides or nucleotide analogs is particularly useful for high-resolution detection of the labeled nucleotides in a single-molecule sequence reaction.
[00142] Importantly, the result of the foregoing treatment is a process for synthesis of nucleic acids, preferably for elucidating a template sequence using substantially only nucleotides, e.g., substantially complete replacement of native nucleotides with nucleotide analogs, and particularly labeled analogs.
Such template dependant synthesis in the presence of substantially only nucleotide analogs, and particularly labeled analogs, also referred to as substantially complete replacement, in sequencing operations is considerably different from previously described sequencing methods where a single nucleotide is substituted with a labeled chain terminating nucleotide among the remaining three natural nucleotides, or where a polymerase template complex are interrogated with only one analog at a time to determine whether such analog is incorporated.
[00143] Another type of suitable nucleotides for the subject sequencing methods allows detection via fluorescence resonance energy transfer (FRET). In FRET, an excited fluorophore (the donor) transfers its excited state energy to a light absorbing molecule (the acceptor) in a distance-dependent manner. The limitation on the distance over which the energy can travel allows one to discern the interactions between labeled molecules and entities in close proximity. Nucleotides of this type can comprise a donor fluorophore attached to the base, ribose or preferably the phosphate backbone (e.g., attached to the terminal phosphate), and an acceptor fluorophore attached to the base, ribose or the phosphate backbone where the donor is not attached. In a preferred embodiment, the donor fluorophore is attached to the terminal phosphate, and an acceptor fluorophore is linked to the base or the ribose unit of the nucleotide. Upon incorporation of this type of nucleotide into the nascent strand, a fluorescent signal can be detected which can be caused by the release of poly-phosphate that is no longer quenched. By determining the order of the fluorescent poly-phosphate that is released upon incorporating a complementary nucleotide during the polymerization event, one can deduce the base sequence of the target nucleic acid. Additional examples of this type of nucleotides is disclosed in U.S. application no. 20030194740.
1001441 In another embodiment, the donor fluorophore can be present in a nucleotide, and the acceptor is located in the polymerase, or vice versa. Where desired, the fluorophore in the polymerase can be provided by a green fluorescent protein (GFP) or a mutant thereof that has a different emission and/or absorption spectrum relative to the wildtype green fluorescent protein. For example, the GFP mutant H9-40 (Tsien et al., Ann. Rev. Biochem. 67: 509 (1998)) which is excited at 399 urn and emits at 511 urn, may serve as a donor fluorophore for use with BODIPY, fluorescein, rhodamine green and Oregon green. In addition, tetramethylrhodamine, LissamineTM, Texas Read and napthofluorescein can be used as acceptor fluorophores with this GFP mutant.
1001451 Other representative donors and acceptors capable of fluorescence energy transfer include, but are not limited to, 4-acetamido-4'-isothiocyanatostilbene-2,2'disulfonic acid;
acridine and derivatives: acridine, acridine isothiocyanate; 5-(2'-aminoethypaminonap- hthalene-l-sulfonic acid (EDANS); 4-amino-N-(3-vinylsulfonyl)phenyl]naphth- alimide-3,5 disulfonate; N-(4-anilino-1-naphthyl)maleimide; anthranilamide;
BODIPY; Brilliant Yellow; coumarin and derivatives: coumarin, 7-amino-4-methylcoumarin (AMC, Cournarin 120), 7-amino-4-trifluoromethylcouluarin (Coumaran 151); cyanine dyes;
cyanosine; 4',6-diaminidino-2-phenylindole (DAPI); 5', 5"-dibromopyrogallol-sulfonaphthalein (Bromopyrogallol Red); 7-diethylamino-3-(4'-isothiocyanatopheny1)4-methylcoumarin; diethylenetriamine pentaacetate; 4,4'-diisothiocyanatodihydro-stilbene-2,-2'-disulfonic acid; 4,4'-diisothiocyanatostilbene-2,2'-disulfonic acid;
54dimethylarninoinaphthalene-1-sulfonyl chloride (DNS, dansylchloride); 4-dimethylarninophenylazopheny14-isothiocyanate (DABITC); eosin and derivatives: eosin, eosin isothiocyanate, erythrosin and derivatives:
erythrosin B, erythrosin, isothiocyanate;
ethidium; fluorescein and derivatives: 5-carboxyfluorescein (FAM), 5-(4,6-dichlorotriazin-2-yl)arnin- fluorescein (DTAF), 2',7'-dimethoxy4'5'-dichloro-6-carboxyfluorescein (JOE), fluorescein, fluorescein isothiocyanate, QFITC, (XRITC); fluorescamine; IR144; IR1446; Malachite Green isothiocyanate; 4-methylumbelliferoneortho cresolphthalein; nitrotyrosine; pararosaniline; Phenol Red; B-phycoerythrin; o-phthaldialdehyde; pyrene and derivative : pyrene, pyrene butyrate, succinirnidyl 1-pyrene; butyrate quantum dots; Reactive Red 4 (Cibacron.TM.
Brilliant Red 3B-A) rhodamine and derivatives: 6-carboxy-X-rhodamine (ROX), 6-carboxyrhodarnine (R6G), lissamine rhodarnine B sulfonyl chloride rhodamine (Rhod), rhodamine B, rhodamine 123, rhodamine X
isothiocyanate, sulforhodarnine B, sulforhodamine 101, sulfonyl chloride derivative of sulforhodarnine 101 (Texas Red); N,N,N,N'-tetramethyl-6-carboxyrhodamine (TAMRA); tetramethyl rhodamine;
tetramethyl rhodamine isothiocyanate (TRITC); riboflavin; rosolic acid; terbium chelate derivatives;
Cy 3; Cy 5; Cy 5.5; Cy 7; IRD 700;
IRD 800; La Jolla Blue; phthalo cyanine; and naphthalo cyanine.
[00146] In alternative configurations, both donor and acceptor fluorophores may be present upon each nucleotide analog, where the donor provides a substantially uniform excitation spectrum, but donates energy to an acceptor that provides an emission spectrum that is different for each type of analog, e.g., A, T, G, or C. Such configurations provide an ability to utilize a single excitation source for multiple different emission profiles, reducing energy input requirements for the systems utilized.
[00147] In addition, xanthene dyes, including fluoresceins and rhodamine dyes can be used as donor and acceptor pairs. Many of these dyes contain modified substituents on their phenyl moieties which can be used as the site for bonding to the terminal phosphate or the base of a nucleotide. Where desired, acceptors acting as quenchers capable of quenching a wide range of wavelengths of fluorescence can be used.
Representative examples of such quenchers include 4-(4'-dimethylaminophenylaz-o)-benzoic acid (DABCYL), dinitrophenyl (DNP) and trinitrophenyl (TNP).
[00148] The polymerization enzymes suitable for the present invention can be any nucleic acid polymerases that are capable of catalyzing template-directed polymerization with reasonable synthesis fidelity. The polymerases can be DNA polymerases or RNA polymerases, a thermostable polymerase or a thermally degradable polymerase wildtype or modified. Non-limiting examples for suitable thermostable polymerases include polymerases from Therm us aquaticus, Therm us caldophilus, Thermus filiformis, Bacillus caldotenax, Bacillus stearothermophus, Therm us thermophilus, Pyrococcus woesei, Pyrococcus furiosus, Thermococcus litoralis, and Thermotoga maritima. Useful thermodegradable polymersases include E. coli DNA polymerase, the Klenow fragment of E. coli DNA polymerase, T4 DNA polymerase, T7 DNA polymerase.
[00149] Additional examples of polymerization enzymes that can be used to determine the sequence of nucleic acid molecules include E. coli T7, T3, SP6 RNA polynierases and AMV, M-MLV and HIV
reverse transcriptases. The polymerase can be bound to the primed target nucleic acid sequence at a primed single-stranded nucleic acid, an origin of replication, a nick or gap in a double-stranded nucleic acid, a secondary structure in a single-stranded nucleic acid, a binding site created by an accessory protein, or a primed single-stranded nucleic acid.
[00150] In one preferred embodiment, the polymerization enzymes exhibit enhanced efficiency as compared to the wildtype enzymes for incorporating unconventional or modified nucleotides, e.g., nucleotides linked with fluorophores. Recombinant DNA techniques can be used to modify the wildtype enzymes. Such techniques typically involve the construction of an expression vector or a library of expression vector, a culture of transformed host cells under such condition such that expression will occur. Selection of the polymerases that are capable of incorporating unconventional or modified nucleotides can be carried out using any conventional sequencing methods as well as the sequencing methods disclosed herein.
1001511 In another preferred embodiment, sequencing is carried out with polymerases exhibiting a high degree of processivity, i.e., the ability to synthesize long stretches of nucleic acid by maintaining a stable nucleic acid/enzyme complex. A processive polymerase can typically synthesize a nascent strand over about 10 kilo bases. With the aid of accessory enzymes (e.g., helicases/primases), some processive polymerases can synthesize even over 50 lcilobases. For instance, it has been shown that T7 DNA polymerase complexed with helicase/primase can synthesize several 100 kilobases of nucleotides while maintaining a stable complex with the target nucleic acid (Kelman et al., "Processivity of DNA Polymerases: Two Mechanisms, One Goal"
Structure 6: 121-125 (1998)).
[00152] In another preferred embodiment, sequencing is performed with polymerases capable of rolling circle replication, i.e., capable of replicating circular DNA templates including but not limited to plasmids. and bacteriophage DNA. A preferred rolling circle polymerase exhibits strand-displacement activity, and preferably has reduced or essentially no 5' to 3' exonuclease activity. Strand displacement results in the synthesis of tandem copies of a circular DNA template, thus allowing re-sequencing the same DNA template more than once. Re-sequencing the same DNA template greatly enhances the chances to detect any errors made by the polymerase, because the same errors unlikely would be repeated by the polymerase and the same error certainly would not be exponentially amplified as in a polymerase chain reaction.
[00153] Non-limiting examples of rolling circle polymerases suitable for the present invention include but are not limited to T5 DNA polymerase (Chatterjee et al., Gene 97:13-19 (1991)), and T4 DNA polymerase holoenzyme (Kaboord and Benkovic, Curr. Biol. 5:149-157 (1995)), phage M2 DNA polymerase (Matsumoto et al., Gene 84:247 (1989)), phage PRD1 DNA polymerase (Jung et al., Proc. Natl. Aced. Sci. USA
84:8287 (1987), and Zhu and Ito, Biochim. Biophys. Acta. 1219:267-276 (1994)), Klenow fragment of DNA
polymerase I (Jacobsen et al., Eur. J.
Biochem. 45:623-627 (1974)).
1001541 A preferred class of rolling circle polymerases utilizes protein priming as a way of initiating replication.
Exemplary polymerases of this class are modified and unmodified DNA
polymerase, chosen or derived from the phages (1329, PRD1, Cp-1, Cp-5, Cp-7, 4)15, 431, 4321, 4)25, BS 32 L17, PZE, PZA, Nf, M2Y (or M2), PR4, PR5, PR722, B103, SF5, GA-1, and related members of the Podoviridae family.
Specifically, the wildtype bacteriophage 4.29 genome consists of a linear double-stranded DNA (dsDNA) of 19,285 base pairs, having a terminal protein vvb 2006/044078 (TP) covalently linked to each 5'end. To initiate replication, a histone-like viral protein forms a nucleoprotein complex with the origins of replication that likely contributes to the unwinding of the double helix at both DNA ends (Serrano etal., The EMBO Journal 16(9): 2519-2527 (1997)). The DNA polymerase catalyses the addition of the first dAMP to the hydroxyl group provided by the TP. This protein-primed event occurs opposite to the second 3' nucleotide of the template, and the initiation product (TP-dA.MP) slides back one position in the DNA to recover the terminal nucleotide After initiation, the same DNA polymerase replicates one of the DNA strands while displacing the other. The high processivity and strand displacement ability of 4)29 DNA
polymerase makes it possible to complete replication of the 4)29 TP-containing genome (TP-DNA) in the absence of any helicase or accessory processivity factors (reviewed by Serrano et al., The EMBO Journal 16(9): 2519-2527 (1997)).
[00155] Modified (1)29 DNA polymerases having reduced 5' to 3' exonuclease activity have also been described (U.S. Patent Nos. 5,198,543 and 5,001,050). These polymerases are particularly desirable for sequencing as the 5' to 3' exonucleases, if present excessively, may degrade the nascent strand being synthesized.
[00156] Strand displacement can be enhanced through the use of a variety of accessory proteins. They include but are not limited to helicases (Siegel et al., J. BioL Chem 267:13629-13635 (1992)), herpes simplex viral protein ICP8 (Skaliter and Lehman, Proc. Natl, Acad. Sci. USA 91(22):10665-10669 (1994)), single-stranded DNA binding proteins (Rigler and Romano, J. Biol. Chem. 270:8910-8919 (1995)), adenovirus DNA-binding protein (Zijderveld and van der Vliet, J. Virology 68(2):1158-1164 (1994)), and BMRF I polymerase accessory subunit (Tsurumi et al., J. Virology 67(12):7648-7653 (1993)).
1001571 In a preferred embodiment, the sequence reaction involves a single complex of strand-displacement polymerization enzyme and a circular target DNA, which is immobilized to an optical confinement. Upon mixing the labeled nucleotides or nucleotide analogs and the primers, the strand-displacement polymerization enzyme directs the synthesis of a nascent strand and a time sequence of incorporating the various types of labeled nucleotides or nucleotide analogs into the nascent strand is registered. Where desired, the strand-displacement polymerase is allowed to synthesize multiple tandem repeats of the target DNA, and thus effecting re-sequencing the same circular DNA target multiple times. It is preferably to register the time sequence of the nucleotides or nucleotide analogs incorporated into at least two tandem repeats of the target DNA molecule, more preferably at least about three to about ten or about three to about one hundred tandem repeats, and preferably no more than about one million repeats. This multiple rounds of or redundant sequencing can take place under an isothermal condition and/or at ambient temperature.

1001581 Using the subject method, sequencing can be carried out at the speed of at least 1 base per second, preferably at least 10 bases per second, more preferably at least 100 bases per second. It has been reported that polymerases can polymerize 1,000 bases per second in vivo and 750 bases per second in vitro (see, e.g. Kelman et al., "Processivity of DNA Polymerases: Two Mechanisms, One Goal," Structure 6:
121-125 (1998); Carter etal., "The Role of Exonuclease and Beta Protein of Phage Lambda in Genetic Recombination. II. Substrate Specificity and the Mode of Action of Lambda Exonuclease," J. Biol. Chem. 246: 2502-2512 (1971); Tabor et al., "Escherichia coli Thioredoxin Confers Processivity on the DNA Polymerase Activity of the Gene 5 Protein of Bacteriophage T7,"
J. Biol. Chem. 262: 16212-16223 (1987); and Kovall et al., "Toroidal Structure of Lambda-Exonuclease" Science 277: 1824-1827 (1997).
Reaction Conditions:
1001591 The sequencing procedures of the present invention are performed under any conditions such that template-directed polymerization can take place using a polymerization enzyme. In one aspect, the substrates of the polymerization enzyme, namely the various types of nucleotides present in the sequence reaction, are adjusted to a physiologically relevant concentration. For example, the nucleotides used in the sequencing reaction are present at a concentration about Michaelis constant of the polymerization enzyme. Such concentration typically ranges from about 1 micromolar to about 50 micromolar or about 100 micromolar.
[00160] The sequencing procedures can also be accomplished using less than four labels employed. With three labels, the sequence can be deduced from sequencing a nucleic acid strand (1) if the fourth base can be detected as a constant dark time delay between the signals of the other labels, or (2) unequivocally by sequencing both nucleic acid strands, because in this case one obtains a positive fluorescence signal from each base pair. Another possible scheme that utilizes two labels is to have one base labeled with one fluorophore and the other three bases with another fluorophore. In this case, the other three bases do not give a sequence, but merely a number of bases that occur between the particular base being identified by the other fluorophore.
By cycling this identifying fluorophore through the different bases in different sequencing reactions, the entire sequence can be deduced from sequential sequencing runs. Extending this scheme of utilizing two labels only, it is even possible to obtain the full sequence by employing only two labelled bases per sequencing run.
[00161] The sequencing procedures can be performed under an isothermal condition, at ambient temperature, or under thermal cycling condition. The choice of buffers, pH and the like is within the skill of practitioners in the art, and hence is not detailed herein.

Detection:
1001621 The subject sequencing method requires the imaging of individual molecules confined in an optical confinement. The polymerase and/or the nucleotides are labeled with fluorophores that emit a distinguishable optical signal when a particular type of nucleotide is incorporated into the nascent strand. The sequence of the distinguishable signals is detected as the nucleotides are sequentially added to the nascent strand within the optical confinement. In a preferred embodiment, such detection is performed without the need to transfer, separation or washing away any reactant or by-product (e.g. fluorophore cleaved from a nucleotide) after each nucleotide addition event. In one aspect of this preferred embodiment, sequence detection is performed without adding reactanti to the mixture prior to reading the next base sequence nucleotide to be incorporated.
[001631 Imaging individual molecules confined in the subject optical confinements is performed with the aid of an optical system. Such system typically comprises at least two elements, namely an excitation source and a photon detector. Numerous examples of these elements are described above.
[00164] In a preferred embodiment, the excitation source is a laser, preferably a polarized laser. The choice of laser light will depend on the fluorophores attached to the different type of nucleotides and/or the polymerases. For most of the flurophorescent compounds, the required excitation light is within the range of about 300 tun to about 700 tun.
For proteinaceous fluorophores such as green-flurorescent protein and mutants thereof, the excitation wavelength may range from about 488 nm to about 404 nm. Those skilled in the art will know or will be able to ascertain the appropriate excitation wavelength to excite a given fluorophore by routine experimentation (see e.g., The Handbook ¨ 'A Guide to Fluorescent Probes and Labeling Technologies, Tenth Edition' (2005) (available from Invitrogen, Inc ./Molecular Probes) 1001651 Another consideration in selecting an excitation source is the choice between one-photon and multiphoton excitation of fluorescence. Multiphoton excitation coupled with detection, also known as multiphoton micropscopy ("MPM"), provides enhanced sensitivity and spatial resolution. MPM is a form of laser-scanning microscopy that uses localized nonlinear excitation to excite fluorescence within a thin raster-scanned plane. In MPM, as in conventional laser-scanning confocal microscopy, a laser is focused and raster-scanned across the sample. The image consists of a matrix of fluorescence intensity measurements made by digitizing the detector signal as the laser sweeps back and forth across the sample. Two-photon excitation probabilities are extremely small, and focusing increases the local intensity at the focal point. Although two-photon excited fluorescence is usually the primary signal source in MPM, three-photon or more excited fluorescence and second or third-harmonic generation can also be used for imaging. See, e.g., a review of multiphoton micropscopy in Webb et al. Nature Biotechnology (2003) 21: (11) 1251-1409. A preferred MPM setup comprises MPM laser scanning microscopes and second-harmonic imaging, equipped with femtosecond mode-locked titanium sapphire lasers operating at wavelengths from about 700 to 1,000 nm. Such setup can capture more than about 100 photons per pixel in most of the conventional imaging multiphoton microscope.
[00166] The sequence of the distinguishable signals can also be detected by other optical systems comprising elements such as optical reader, high-efficiency photon detection system, photo multiplier tube, gate sensitive FET's, nano-tube FET's, photodiode (e.g. avalanche photo diodes (APD)), camera, charge couple device (CCD), electron-multiplying charge-coupled device (EMCCD), intensified charge coupled device (ICCD), and confocal microscope.
[00167] A preferred combination comprises wide field CCD or ICCD and intensified video imaging microscopes with digital image processing capability, as well as Fluorescence Photobleaching Recovery (FPR) and Fluorescence Correlation Spectroscopy (FCS) coupled with confocal multiphoton capability and continuous data acquisition and control. Such set up may further comprise modular instrument for quasi-elastic light scattering, laser DIC
interferometry, correlation spectroscopy instrumentation, components of optical force microscopy, and Time Correlated Single Photon Counting (TCSPC).
[00168] These optical systems may also comprise optical transmission elements such as diffraction gratings, arrayed waveguide gratings (AWG), optic fibers, optical switches, mirrors, lenses (including microlens and nanolens), collimators. Other examples include optical attenuators, polarization filters (e.g., dichroic filter), wavelength filters (low-pass, band-pass, or high-pass), wave-plates, and delay lines. In some embodiments, the optical transmission element can be planar waveguides in optical communication with the arrayed optical confinements.
[00169] These and other optical components known in the art can be combined and assembled in a variety of ways to effect detection of the distinguishable signals emitted from the sequencing reaction. Preferred devices allow parallel data collection using arrays having a large number of optical confinements, where simultaneous and independent sequencing of nucleic acids takes place. In one aspect, the preferred system can collect and process signals from more than 104 optical confinements, more than 2x104 optical confinements, or more than 105 optical confinements, or more than 2x105 optical confinements, or preferably more than 106, or preferably more than 2x106 optical confinements, and even more preferably more than 107 or 2 x 10 opticalconfinements. In another aspect, the preferred setup can monitor in real time the simultaneous and independent sequencing of nucleic acids at a speed ot about I base per second, preferably at a speed of about 10 bases per second, more preferably at a speed of about 100 bases per second and even more preferably at 1,000 bases per second. As such, the massive parallelism coupled with the rapid sequencing reaction can provide an overall sequencing output greater than 100,000 bases per second.
The overall output can be scaled up to at least 1 megabase per second, preferably 10 or more megabases per second.
Further by obtaining such date from multiple different sequence fragments e.g., in from two or more different reaction volumes, one can obtain independent sequences, e.g. from contiguous fragments of genomic DNA, allowing the high rate of throughput that is directly applicable to genomic sequencing.
Other Single-Molecule Applications:
1001701 The subject optical confinements and arrays of optical confinements find utility in many other chemical and biological applications where single molecule analyses are desired. In general, the subject optical confinements are applicable for any single molecule analysis involving any reagent that can be attached to the surface and for which substrates can be labeled, including, enzymes, nucleic acids, antibodies, antigens, and the like. Such applications include discerning interactions involving biological molecules such as proteins, glycoproteins, nucleic acids, and lipids, as well as inorganic chemicals, or any combinations thereof The interactions may be between nucleic acid molecules, between nucleic acid and protein, and between protein and small molecules.
[00171] Abnormalities in interactions involving biological molecules have long been acknowledged to account for a vast number of diseases including, numerous forms of cancer, vascular diseases, neuronal, and endocrine diseases.
An abnormal interaction, in form of e.g., constitutive activation and premature inactivation of a signaling complex, are now known to lead to aberrant behavior of a disease cell. In the case of cancer, abnormal interactions between two signaling transduction molecules, such as growth factor receptors and their corresponding ligands, may result in dysfunction of cellular processes, which ultimately lead to dysregulated growth, lack of anchorage inhibition, genomic instability and/or propensity for cell metastasis.
[00172] A specific interaction between biological or chemical molecules typically involves a target molecule that is being investigated and a probe suspected to be able to specifically interact with the target. In practicing the subject methods, the target and the probe are placed within an optical confinement.
The target-probe complex can be a protein-protein complex, a glycoprotein-protein complex (e.g., receptor and ligand complex), a protein-nucleic acid complex (e.g., transcription factor and nucleic acid complex), a protein-lipid complex, and complex of inorganic or organic small molecules.

[00173] Preferably, each optical confinement contains only one target that is being investigated. This can be achieved by diluting a minute amount of target in a large volume of solution, such that deposition over an array of confinements results in a primary distribution, or a majority of confinements will have a single target molecule disposed there. Alternatively, a non-cylindrical waveguide, wherein the opening of the waveguide core is narrower in lateral dimension than the base, can be used to restrict the entry of multiple target proteins while permitting the entry of a number of smaller probes.
[00174] The target or probe can be immobilized onto the inner surface of the optical confinement by any of the methods applicable for immobilizing and depositing the polymerases described in the section above. Such methods encompass the uses of covalent and noncovalent attachments effected by a variety of binding moieties. The choice of the binding moieties will depend on the nature of the target and/or the probe. For example, the binding moieties can be synthetically linked to the protein target or the probe, or made as a fusion motif or tag via a recombinant means. A preferred way to immobilize the target protein or the proteinaceous probe involves the use of the streptavidin or avidinibiotin binding pair, and any other binding moieties or agents described above.
[00175] The reaction conditions will depend on the particular interaction that is under investigation. One may vary the reaction temperature, the duration of the reaction, the buffer strength, and the target concentration or the probe concentration. For example, one may vary the concentration of the probe in order to measure its binding affinity to the target protein. To determine the thermal stability of the target-probe complex, one may vary the reaction temperature. Stability of the target-probe complex can also be determined by varying the pH, or buffer salt concentration. Where desired, the interaction can be studied under physiologically relevant temperature and buffer conditions. A physiologically relevant temperature ranges from approximately room temperature to approximately 37 C. A physiological buffer contains a physiological concentration of salt at neutral pH ranging from about 6.5 to about 7.8, and preferably from about 7.0 to about 7.5. Adjusting the reaction conditions to discern a particular interaction in vitro between a given target and a probe is within the skill of artisans in the field, and hence is not detailed herein.
[001761 The target and/or the probe are generally labeled with detectable labels so that a photon detector can detect a signal indicative of their interaction. Suitable labels encompass all of those labels disclosed in the Single-Molecule Sequencing section. Preferred labels are luminescent labels, and especially fluorescent or chromogenic labels.
[001771 In one embodiment, the target is labeled with a fluorophore whose signal is quenched upon interaction with the corresponding probe conjugated with an appropriate quencher. A variety of suitable fluorophore-quencher pairs is disclosed in the section above and hence is not detailed herein. A
variation of this embodiment is to label the target and the probe with donor and acceptor fluorphores (or vise versa) that emit a distinguishable signal when the two molecules bind to each other. A wide range of applicable donor and acceptor fluorophores is also described above. Those of skill in the art will appreciate the wide diversity of detectable labels and the combinations thereof to generate a distinguishable signal that is indicative of a specific interaction between biological molecules and/or chemical compounds.
[00178] The detection of the distinguishable signal indicative of a specific interaction is performed with the aid of the optical systems described herein. Any of the systems applicable for single-molecule sequencing is equally suited for detecting interactions between other biological molecules and/or chemical compounds. A preferred system allows parallel data collection using arrays having a large number of optical confinements, where simultaneous and independent target-probe interactions can take place. In one aspect, the preferred system can collect and process signals from more than 104 optical confinements, more than 2x104 optical confinements, more than 105 optical confinements, more than 2x105 optical confinements, preferably more than 106, or preferably more than 2x106 optical confinements, and even more preferably more than 107 or 2x107 optical confinements.
[00179] Of particular significance is the application of the aforementioned method in detecting the presence of a specific protein-protein interaction. Such application generally employs a proteinaceous probe and a target protein placed in an optical confinement. In one aspect of this embodiment, the specific protein-protein interaction is between a cell surface receptor and its corresponding ligand. Cell surface receptors are molecules anchored on or inserted into the cell plasma membrane. They constitute a large family of proteins, glycoproteins, polysaccharides and lipids, which serve not only as structural constituents of the plasma membrane, but also as regulatory elements governing a variety of biological functions. In another aspect, the specific protein-protein interaction involves a cell surface receptor and an immunoliposome or an immunotoxin. In yet another aspect, the specific protein-protein interaction may involve a cytosolic protein, a nuclear protein, a chaperon protein, or proteins anchored on other intracellular membranous structures. In yet another aspect, the specific protein-protein interaction is between a target protein (e.g., an antigen) and an antibody specific for that antigen.
[001801 The specific interaction between an antigen and an antibody has been explored in the context of immunoassays. There exists a variety of immunoassays in the art, but none of which permits single-molecule detection. For instance, the conventional radioimmunoassay detects the interactions between a population of antigens and a population of radioactively labeled antibodies on an immunoblot. Another conventional immunoassay termed ELISA (Enzyme Llinked Immunoradiometric Assay) utilizes an antigen-specific antibody and an enzyme-lined generic antibody that binds to the specific antibody. The specific interaction between the antigen and the antibody is visualized upon addition of the substrate to the linked enzyme. Such assay again is performed on an immunoblot providing an ensemble measurement of all interactions detected.
[001811 The subject optical confinement provides an effective tool for conducting a single- molecule immunoassay.
Unlike the conventional immunoassays, the specific interaction between the antigen and the antibody can be resolved at the single-molecule level. While all of the optical confinements embodied in the present invention are applicable for conducting single-molecule immunoassays, a particularly desirable system comprises an array of optical confinements with a relatively high fill fraction ratio. For example, a preferred system comprises an array of waveguides having a fill fraction greater than 0.0001, more preferably greater than about 0.001, more preferably greater than about 0.01, and even more preferably greater than 0.1.
1001821 In practicing the subject immunoassays, the antibodies an be labeled with a suitable label selected from radioactive labels, fluorescent labels, chemiluminescent labels, enzyme tags, such as digoxigenin, B-galactosidase, urease, alkaline phosphatase or peroxidase, avidin/biotin complex, and any of the detectable labels disclosed herein.
[001831 The subject immunoassays can be performed to characterize biological entities, screen for antibody therapeutics, and determine the structural conformations of a target antigen.
For instance, immuoassays involving antibodies that are specific for the biological entity or specific for a by-product produced by the biological entity have been routinely used to identify the entity by forming an antibody-entity complex. Immunoassays are also employed to screen for antibodies capable of activating or down-regulating the biological activity of a target antigen of therapeutic potential. Immunoassays are also useful for determining structural conformations by using anti-idotypic antibodies capable of differentiating target proteins folded in different conformations.
[00184] Another important application of the aforementioned single-molecule analysis is to study enzyme kinetics, which may include determining the enzymatic turnover cycle, the dynamic behavior, folding and unfolding intermediates, and binding affinities. The enzymes under investigation may be immobilized within the optical confinements or present in solutions confined within the subject optical confinements.
[00185] All of the optical confinements embodied by the present invention can be employed to study enzyme kinetics. The choice of a specific optical confinement will depend on the specific characteristic that is under investigation. For instance, an optical confinement comprising a non-cylindrical core having an opening on the upper surface that is narrower than that of the base of the optical confinement is preferable for measuring the association rate constant (on-rate) of an enzymatic reaction. This configuration significantly restricts the diffusion of reactants or substrates, and hence increases the average residence time in the observation volume. On the other nana, an optical confinement comprising a core with an opening that is wider in lateral dimension than the base imposes impose a stearic or entropic hinderence to entering the structure, hence is useful for measuring the accessibility for large enzymes or enzymatic complexes.
Uses of the Subject Optical Confinements in Ensemble Measurements:
[00186] While the optical confinements of the present invention are particularly useful in conducting single-molecule analyses, the subject confinements are also suited for high throughput performance of ensemble bulk measurements. Accordingly, the present invention provides a method of detecting interactions among a plurality of molecules, comprising: placing said plurality of molecules in close proximity to an array of zero-mode waveguides, wherein individual waveguides in said array are separated by a distance sufficient to yield a detectable intensity of diffractive scattering at multiple diffracted orders upon illuminating said array with an incident wavelength of light beam; illuminating said array of zero-mode waveguides with said incident wavelength; and detecting a change in said intensity of diffractive scattering of said incident wavelength at said multiple diffracted orders, thereby detecting said interactions among said plurality of molecules.
[00187] Arrays employed for this method typically comprises optical confinements spaced far apart relative to the - s incident wavelength. Such spacing of the individual optical confinements far apart relative to the illuminating radiation (e.g., half of the wavelength of the illuminating radiation) creates a larger effect on the diffractive scattering of incident light at a given angle away from the angle of specula reflection. In one aspect of this embodiment, the arrays contain individual confinements separated by more than one wavelength of the incident radiation, usually more than 1.5 times the incident wavelength, but usually does not exceed 150 times the incident wavelength.
[00188] Arrays having the optical confinements spaced far apart relative to the incident wavelength also have desirable properties. While the angle-dependent scattering may raise the background signal that could be disadvantageous for certain applications, it provides a means particularly suited for characterizing the size and shape of the optical confinements. It also readily permits ensemble bulk measurements of molecule interactions, involving especially unlabelled molecules. Arrays suited for such applications generally contain individual confmements separated by more than one wavelength of the incident radiation, usually more than 1.5 times the incident wavelength, but usually not exceeding 150 times the incident wavelength.
[00189] The ensemble bulk measurement is typically performed with the aid of the optical systems described herein. Any of the setup applicable for single-molecule sequencing is equally suited for this analysis.

[00190] Further illustrations of the fabrication of the optical confinements of the present invention and then uses in sequencing are provided in the Example section below. The examples are provided as a guide to a practitioner of ordinary skill in the art, and are not meant to be limiting in any way.
EXAMPLES

[00191] The following provides an illustrative process of fabricating zero-mode waveguide. The parameters described herein are meant to be illustrative and not intended to be limiting in any manner.
1. Substrates: Substrates are double polished, 60/40 scratch/dig surface quality, Fused Silica wafers, cut to 100millimeters(+/- 0.2mm) diameter, and 175micrometer(+/- 25 micrometers) thick and a total thickness variation of less than 25 micrometers.
2. Clean: A mix of 5 parts deionized water, 1 part of (30% v/v Hydrogen Peroxide in water), 1 part of (30% v/v Ammonium Hydroxide in water) is heated to 75 degree Celsius on a hotplate. The wafers are immersed in the mix using a Teflon holder or other chemically resistant holder for a duration of 15 minutes.
3. Rinsing: The holder containing the wafers is removed from the RCA clean bath and immersed in a bath of deionized water. The wafers are left in this second bath for a 2minutes period. The holder still containing the wafers is removed from the bath, and sprayed with deionized water to thoroughly finish the rinsing process.
4. Drying: Within a minute of the final rinsing step, the wafers are dried, while still in the holder, using a dry clean nitrogen flow.
5. Oxygen Plasma: The wafers are then placed in a Glenn 1000p plasma Asher, used in plasma etch mode (wafers on a powered shelf, and under another powered shelf), with 140mTorr pressure and 400Watts of forward power at 40kHz frequency. The plasma is maintained for 10 minutes. A flow of 18sccm of molecular oxygen is used.
6. Vapor Priming: The wafers are loaded within 3 minutes after the Oxygen plasma in a Yield Engineering Systems vapor priming oven where they are coated with a layer of HexaMethylDiSilazane (HMDS) adhesion promoter.
7. Electron beam resist coating: The wafers are coated within 15 minutes after the Vapor Priming in a manual spinner unit using NEB-31 electron beam resist (Sumitomo Chemical America). About 3m1 are dispensed on the wafer, which is then spun at 4500 rpm for 60 seconds. Initial acceleration and deceleration are set to 3 seconds 8. Resist Bake: The wafers are baked on a CEE hotplate at a temperature of 115 degree Celsius for 2 minutes. The plate is equipped with a vacuum mechanism that allows good thermal contact between the wafers and the hotplate surface.
9. Gold Evaporation: a layer of lOnm of gold is then thermally evaporated on the Wafers, on the side coated with the resist. A pressure of less than 2 10e-06 TOIT must be reached before the evaporation. The evaporation is performed at a rate of approximately 2.5 Angstrom per second and monitored using an Inficon controller.
10. Electron beam exposure: a pattern consisting of Zero Mode Waveguides is exposed on the wafers, using a high resolution electron beam lithography tool such as a Leica system. Zero mode waveguides are patterned as single exel features. At a current of nominally 1 nanoAmpere, and a Variable Resolution Unit of 1, and for an exel setting of 5 nanometers, doses can range from 10000 microCoulombs per square centimeters to 300000 microCoulombs per square centimeters.
11. Post Exposure Bake: The wafers are then submitted to a 2 minute post exposure bake on a hotplate at 95 degree Celsius, equally equipped with a vacuum mechanism.
12. Gold Etch: After removal from the electron beam system, the lOnanometer gold layer is removed using gold etchant TFA at room temperature (GE 8148, Transene Corporation), for 10 seconds. Wafers are held in a Teflon holder similar to the one used in step 2.
13. Rinsing: The holder containing the wafers is removed from the gold etchant bath and immerse in a bath of deionized water. The wafers are left in this second bath for a 2 minutes period or shorter with gentle manual agitation. The holder still containing the wafers is removed from the bath, and sprayed with deionized water to thoroughly finish the rinsing process. Alternatively, the holder still containing the wafer is then placed into a new container containing fresh deionized water.
14. Drying: Within a minute of the final rinsing step, the wafers are dried, while still in the holder, using dry clean nitrogen flow.
15. Post Exposure Bake: The wafers are then submitted to a 2 minute post exposure bake on a hotplate at 95 degree Celsius, equally equipped with a vacuum mechanism.
53 =
16. Developing: The wafers still in the chemically resistant holder are inunersed in developer MF-321 (Shipley Chemicals, Rohm-Haas) at room temperature for duration of 30 seconds.
17. Rinsing: The holder containing the wafers is removed from the developer etchant bath and immerse in a bath of deionized water. The wafers are left in this second bath for a 2 minutes period with gentle manual agitation. The holder still containing the wafers is removed from the bath, and sprayed with deionized water to thoroughly finish the rinsing process.
18. Drying: Within a minute of the final rinsing step, the wafers are dried, while still in the holder, using dry clean nitrogen flow.
19. Surface Descum: The wafers are loaded in a Glenn 1000p plasma asher run in ashing mode (Wafers on a grounded plate below a powered plate), and submitted to a 30 seconds surface descuming oxygen plasma at a pressure of 140 mTorr and a power of 100 Watts forward power at 40kHz. A flow of 18sccm of molecular oxygen is used.
20. Aluminium Evaporation: The wafers are loading in a metal evaporator within 5 minutes of the surface descum process. A layer of 100nm of thermally evaporated Aluminium is now deposited on the wafers. Evaporation is made at a pressure of no less than 2 10^-6 Ton at a rate of 25 Angstrom per seconds and monitored using an Inficon controller.
21. Aluminium Thickness measurement: The thickness of the aluminium is measured using a P-10 Profilometer (Tencor).
22. Zero Mode Waveguide Decasting: The Zero Mode Waveguide are decasted from the enclosing Aluminium film by immersing them, in a Teflon holder or other chemically resistant holder, in a bath of 1165 Stripper (Shipley Chemicals, Rohm-Haas), or in a bath of AZ-300T Stripper (Shipley Chemicals, Rohm-Haas). The bath is submitted to sonication by immersing the Container holding both the Stripper and the wafer holder in a sonicator. The wafers are left in the decasting bath for 30 minutes or longer for about 45 minutes, and are provided with additional gentle agitation.
23. Rinsing: The stripping bath is removed from the sonicator. The wafers are removed from the stripper bath and immerse in a bath of deionized water. The wafers are left in this second bath for a 2 minutes period with gentle manual agitation. The wafers are removed from the bath, and sprayed with deionized water to thoroughly finish the rinsing process.
24. Drying: Within a minute of the final rinsing step, the wafers are dried, while still in the holder, using dry clean nitrogen flow 25. Photoresist coating: The wafers are coated with Shipley 1827 photoresist spun at a speed of 1500 rpm. About 5m1 of resist is dispensed. Acceleration and deceleration is set to 5 seconds.
26. Resist Bake: The wafers are baked on a CEE hotplate at a temperature of 115 degree Celsius for 15 minutes. The plate is equipped with a vacuum mechanism that allows good thermal contact between the wafers and the hotplate surface.
27. Dicing: The wafer are diced using a K&S-7100 dicing saw (Kulicke & Soffa) using a resin/diamond blade (ADT 00777-1030-010-QIP 600). The wafers are mounted on a low-tack adhesive tape prior to dicing.
28. Die Removal: The dies are removed from the adhesive tape manually and stored.
29. Resist removal: The layer of 1827 photoresist is removed by immersing the dies first in an acetone bath for 1 minute, then in a 2-propanol bath for 2 minute with gentle manual agitation.
30. Die Drying: The die is dried after being removed from the 2-propanol bath using dry clean air.
31. Plasma Clean: The wafers are loaded in a Drytek 100 plasma etcher, and submitted to a 1 minute oxygen plasma at a pressure of 140 mTorr, a molecular oxygen flow of 85 sccm oxygen and an RF power of 500 Watts forward power at 13Mhz. Alternatively, Harrick Plasma Cleaner PDC-32G are submitted to a 5 minute dry clean air plasma at a pressure of 2 Torr and 10.5 Watts of power.

Monitoring enzymatic synthesis of a DNA strand by a single DNA polymerase molecule in real time [00192] This experiment can be performed using the optical system and reaction mixtures detailed below.
However, the reference to any particular optical system and parameter, buffer, reagent, concentration, pH, temperature, or the like, is not intended to be limiting. They are included to provide as one illustrative example of carrying out the methods of the present invention.
[00193] Enzymatic synthesis of a DNA strand by a single DNA polymerase molecule was tracked in real time using a fluorescently labeled nucleotides. Individual Phi29N62 DNA polymerase enzymes (Amersham Biosciences, Piscataway, NJ) were immobilized in zero-mode waveguides (ZMWs) by non-specific binding using a dilute enzyme solution. After immobilization, the ZMW structures were washed to remove unbound enzyme, and then exposed to a solution containing the reaction reagents. As of the DNA
template, a 70-bp pre-primed circular DNA
sequence was used that contained two guanine bases in characteristic, asymmetric spacing (Figure 9A). Strand-displacement polymerizing enzymes such as Phi29 DNA polymerase will continuously loop around the circular template and thus generate a long and highly repetitive complementary DNA
strand.
1001941 An R110-dCTP (Amersham Biosciences, Piscataway, NJ) was used as the fluorescently-tagged nucleotide analog in which the fluorophore is attached to the nucleotide via a linker to the gamma-phosphate. In contrast to the more commonly used base-labeled nucleotide analogs, gamma-phosphate-linked analogs are cleaved through the enzymatic activity of DNA polymerase as the attached nucleotide is incorporated into the growing DNA strand and the label is then free to diffuse out of the effective observation volume surrounding the DNA polymerase. The efficient removal of the fluorophore ensures continuously low background levels and prevents significant interference with DNA polymerase activity. These features of the gamma-phosphate-linked fluorophore are preferable for this application because they will enable replacement of all four bases with fluorophore-tagged analogs. Binding of a nucleotide and its subsequent incorporation into nucleic acid from a mismatch event is distinguished because the rate constants of these two processes are significantly different, and because nucleotide incorporation involves several successive steps that prevent zero delay time events.
1001951 All other nucleotides were supplied without labels. We have established a very effective way of removing any remaining trace amount of native dNTP in a nucleotide analog preparation to ensure that errors are not introduced due to the incorporation of unlabeled dNTPs by an enzymatic purification using an alkaline phosphatase prior to the polymerization assay.
1001961 To investigate the speed and processivity of the Phi29N62 DNA
polymerase under these conditions, the incorporation characteristics were measured using R110-dCTP completely replacing dCTP in the reaction mixture, both in solution and with enzyme immobilized on a glass surface. It was found that the enzyme efficiently utilized this analog, synthesizing complementary DNA of many thousands of base pairs in length without interruption in a rolling circle synthesis protocol, using both small preformed replication forks (Figure 9A) as well as larger circular DNA such as MI3 DNA. Only two, asymmetrically spaced R110-dCTPs were to be incorporated into this template.
Similar experiments demonstrated that DNA polymerase can be immobilized to the bottom of ZMWs without losing this catalytic activity.

[00197] The incorporation of the fluorescently labeled dCTP nucleotide was tracked during rolling-circle DNA
synthesis by recording the fluorescent light bursts emitted in an individual ZMW. DNA polymerase activity was observed in many waveguides as distinct bursts of fluorescence, which lasted several minutes. The fluorescence time trace showed a characteristic double burst pattern (Figure 9B), each burst corresponding to an incorporation event of a R110-dCTP analog into the DNA strand and subsequent cleavage of the fluorophore. In histograms of burst intervals derived from the full time trace, two peaks corresponding to DNA synthesis along the short (14 bases, approximately one second) and long (54 bases, approximately four seconds) DNA
template segments are visible, consistent with an overall average speed measured in bulk solution under these conditions of approximately ten base pairs per second.
[001981 It is noteworthy that this single-molecule activity at a fluorophore concentration of 10 uM was readily observable. In conventionally created excitation volumes, the number of fluorophores would be far too high to permit the observation of individual enzymatic turnovers of DNA polymerase.
These experiments thus confirmed the validity of the ZMW-based single-molecule DNA sequencing approach by verifying that (a) immobilization of DNA polymerase in ZMWs does not affect its enzymatic activity; (b) fluorescent gamma-phosphate-linked nucleotide analogs do not inhibit the activity of DNA polymerase; and (c) ZMWs provide an adequate degree of confinement to detect single-molecule DNA polymerase activity at physiological concentrations of reagents. More generally, these results prove that ZMWs allow single-molecule analysis of enzyme kinetics, especially involving any enzyme that can be attached to the surface and for which substrates can be fluorescently labeled.

Real time sequencing using multiple different labeled nucleotides [001991 An experiment similar to that described in Example 2, above, was performed using two different labeled nucleotide analogs. The experiment can be performed using the optical setup or system and reaction mixtures detailed as follows. However, the reference to any particular optical setup and parameter, buffer, reagent, concentration, pH, temperature, or the like, is not intended to be limiting.
They are included to provide as one illustrative example of carrying out the methods of the present invention.
[002001 Preparing reaction samples: Approximately 10121 of reaction mixture is used in one sequencing reaction.
The reaction mixture generally contains 0.5-1 mM MnC12, 0.1-1 uM DNA template, 10 uM dATP, 10 tiM dGTP, 10 uM SAP-treated Alexa 488-dC4P, and 10 uM SAP-treated Alexa 568-dT4P, and DNA
polymerase. The labeled dC4P and dT4P can also be substituted with labeled dA4P and dG4P.

1002011 Preparing Zero-mode waveguide: Prior to the polymerization reaction, a zero-mode waveguide is typically refreshed in a plasma cleaner. A PDMS gasket covering the ZMW is placed onto the waveguide to cover the individual optical confinements. An aliquot of the reaction mixture described above except the DNA polymerase is applied without touching the waveguide surface. The diffusion background is measured. If the background (i.e., fluorescence burst from the ZMW) low and acceptable, then DNA polymerase will be applied to ZMW and immobilized thereon. The immobilization mixture typically contains 0.5 to 1 mM
MnC12, 0.1 to luM template, 15 nM DNA polymerase, in a buffer of 25mM Tris-HCL, pH 7.5 and lOrnM beta-mercaptoethanol. The polymerase is allowed to stick to the surface of ZMW after an incubation of about 15 minutes at about 0 C. The immobilization reaction mixture is then removed, replaced with the reaction mixture described above.
1002021 A microscope system equipped with an appropriate laser, e.g., Ar/Kr laser, is used that includes an optical setup for simultaneous collection and detection of signals from multiple different waveguides, and for the resolution of each of the A488 and 568 fluorophores present. The system includes an objective lens and a series of dichroics/notch-off filters for separating emitted fluorescence from reflected excitation light. The emitted signals are passed through a wedge filter to spatially separate the signal component of each fluorophore, and each signal is imaged onto an EMCCD camera.
[002031 Polymerase Activity Measurement: The ZMW is placed under the microscope. Polymerization reaction is then monitored using a camera for a desired period of time, e.g., two minutes or longer, after the transillumination light is applied. The data is automatically transmitted to a computer that stores and trace the fluorescence burst of each reaction in the ZMW.
1002041 A circular DNA having either a block of repeating A bases followed by a block of G bases, or a series of repeating A-G bases was sequenced according to the aforementioned procedures.
A representative profile of the fluorescence bursts corresponding to each incorporation event of the labeled nucleotides is depicted in Figure 16, which indicates that real-time and single-molecule sequencing has been achieved with more than one type of labeled nucleotides. Statistical analysis of pulse data from multiple separate repeats and multiple different waveguides establishes the sequence dependant detection of incorporation of labeled bases in real time.
1002051 Although described in some detail for purposes of illustration, it will be readily appreciated that a number of variations known or appreciated by those of skill in the art may be practiced within the scope of present invention.

Claims (186)

1. An apparatus comprising:
an array of optical confinements having a density on a substrate exceeding 4x10 4 confinements per mm2, wherein said optical confinements are suitable for holding a biological reagent, and wherein said optical confinements provide an effective observation volume that permits observation of individual molecules present in said biological reagent; and an optical system operatively coupled to the optical confinements that detects signals from the effective observation volume of said confinements.
2. The apparatus of claim 1, wherein said density exceeds 10 5 confinements per mm2.
3. The apparatus of claim 1 or 2, wherein said optical confinements hold a biological reagent.
4. The apparatus of claim 3, wherein said biological reagent comprises an enzyme.
5. The apparatus of claim 3, wherein said biological reagent further comprises substrates of said enzyme present at a concentration of the Michaelis constant (Km) of said enzyme for said substrates.
6. The apparatus of claim 5, wherein said concentration is higher than 1 micromolar.
7. The apparatus of claim 5, wherein said concentration is higher than 50 micromolar.
8. The apparatus of claim 5, wherein said substrates are labeled.
9. The apparatus of claim 5, wherein said individual molecules are products of a reaction between said enzyme and said substrates contained in said biological reagent.
10. The apparatus of claim 3, wherein said individual molecules are DNA/polymerase complexes.
11. The apparatus of claim 3, wherein said individual molecules are biomolecules selected from the group consisting of nucleic acids, polypeptides, and combinations thereof
12. The apparatus of claim 3, wherein said individual molecules are organic or inorganic chemical compounds.
13. The apparatus of claim 3, wherein at least one individual confinement is separated from at least one other optical confinement in said array by a distance less than 1000 nm.
14. The apparatus of claim 3, wherein said optical confinements in said array are separated from each other by a distance between 200 nm and 1000 nm.
15. The apparatus of claim 3, wherein said array comprises at least 2x105 optical confinements.
16. The apparatus of claim 3, wherein said array comprises at least 107 optical confinements.
17. The apparatus of claim 3, wherein said individual molecules are labeled with a detectable label.
18. The apparatus of claim 17, wherein said detectable label is selected from the group consisting of a fluorescent label, a chemiluminescent label, and a radioactive label.
19. The apparatus of claim 3, wherein said optical confinements are made of porous film.
20. The apparatus of claim 3, wherein each individual confinement in said array is operatively coupled to an optical transmission element transmitting an incident light.
21. The apparatus of claim 3, wherein said optical system further comprises a photon detector.
22. The apparatus of claim 3, wherein said optical confinements are waveguides comprising a cladding surrounding a core, wherein said cladding precludes propagation of a majority of incident light having a wavelength greater than a cutoff wavelength through said core.
23. The apparatus of claim 22, wherein said waveguides preclude propagation of more than 90% of incident light having a wavelength greater than a cutoff wavelength through said core.
24. The apparatus of claim 22, wherein said waveguides preclude propagation of more than 99% of incident light having a wavelength greater than a cutoff wavelength through said core.
25. The apparatus of claim 22, wherein said cladding comprises a sheet, and one or more holes extending through said sheet where each hole constitutes the core.
26. The apparatus of claim 22, wherein said core is cylindrical in shape.
27. The apparatus of claim 22, wherein said core is non-cylindrical in shape.
28. The apparatus of claim 22, wherein said cladding comprises a metal alloy.
29. The apparatus of claim 28, wherein said alloy is aluminum alloy.
30. The apparatus of claim 3, wherein each individual confinement provides an effective observation volume less than 1000 zeptoliters.
31. The apparatus of claim 3, wherein each individual confinement provides an effective observation volume less than 80 zeptoliters.
32. The apparatus of claim 3, wherein each individual confinement provides an effective observation volume less than 10 zeptoliters.
33. A kit comprising an array of optical confinements of claim 3 and an instruction manual for use of said array.
34. The apparatus of claim 3, wherein a plurality of said optical confinements in said array each holds a single complex of a target nucleic acid and a polymerization enzyme, and more than one type of labeled nucleotides or nucleotide analogs present at a concentration 1 uM to 50 uM.
35. The apparatus of claim 34, wherein said optical confinements are zero-mode waveguides.
36. A method of performing multiple chemical reactions involving a plurality of reaction samples, comprising:
(a) providing an array of optical confinements of claim 1;
(b) placing the plurality of reaction samples comprising labeled reactants into the optical confinements in the array, wherein a separate reaction sample is placed into a different confinement in the array;
(c) subjecting the array to conditions suitable for formation of products of the chemical reactions;
and (d) detecting the formation of the products with said optical system.
37. The method of claim 36, wherein the step of detecting comprises illuminating the different confinements with an incident light beam and detecting an optical signal emitted from the reaction samples.
38. The method of claim 36, wherein the chemical reactions involve protein-protein interactions.
39. The method of claim 36, wherein the chemical reactions involve nucleic acid-protein interactions.
40. The method of claim 36, wherein the chemical reactions involve nucleic acid-nucleic acid interactions.
41. A method of detecting a biological analyte, comprising:
optically capturing the analyte within an optical confinement that is created by (a) providing an array of optical confinements having a density on a substrate exceeding 4x10 4 confinements per mm2, wherein said optical confinements provide an effective observation volume that permits observation of individual molecules; and an optical system operatively coupled to the optical confinements that detects signals from the effective observation volume of said confinements; and (b) illuminating at least one optical confinement within the array that is suspected to contain the analyte with an incident light beam thereby detecting the analyte.
42. The method of claim 41, wherein said density on a substrate exceeding 10 5 confinements per mm2.
43. The method of claim 41, wherein said analyte is labeled with a detectable label.
44. The method of claim 41, wherein said analyte is selected from the group consisting of nucleic acid, polypeptide, organic compound, and inorganic compound.
45. A method of sequencing a plurality of target nucleic acid molecules, comprising:
(a) providing an array of optical confinements having a density on a substrate exceeding 4x10 4 confinements per mm2, wherein said optical confinements provide an effective observation volume that permits observation of individual molecules; and an optical system operatively coupled to the optical confinements that detects signals from the effective observation volume of said confinement;

(b) mixing in the optical confinements the plurality of target nucleic acid molecules, primers complementary to the target nucleic acid molecules, polymerization enzymes, and more than one type of nucleotides or nucleotide analogs to be incorporated into a plurality of nascent nucleotide strands, each strand being complementary to a respective target nucleic acid molecule;
(c) subjecting the mixture of step (b) to a polymerization reaction under conditions suitable for formation of the nascent nucleotide strands by template-directed polymerization of the nucleotides or nucleotide analogs;
(d) illuminating the optical confinements with an incident light beam; and (e) identifying the nucleotides or the nucleotide analogs incorporated into the each nascent nucleotide strand.
46. The method of claim 45, wherein the step of identifying yields a time sequence of incorporation of the nucleotides provided in step (b).
47. The method of claim 45, wherein the identifying step is effected while the template-directed polymerization is taking place.
48. The method of claim 45, wherein the target nucleic acid molecules are selected from the group consisting of circular DNA molecules, linear DNA molecules, RNA
molecules, and DNA/RNA hybrids.
49. An apparatus comprising: an array of waveguides on a solid support, said array having a fill fraction greater than 0.0001, wherein said waveguides are suitable for holding a biological reagent, and wherein said waveguides provide an effective observation volume that permits observation of individual molecules present in said biological reagent; and an optical system operatively coupled to the waveguides that detects signals from the effective observation volume of said waveguides.
50. The apparatus of claim 49, wherein said array has a fill fraction greater than 0.001.
51. The apparatus of claim 49, wherein said array has a fill fraction greater than 0.01.
52. The apparatus of claim 49, wherein said array has a fill fraction greater than 0.1.
53. The apparatus of claim 49, wherein said array has a fill fraction ranging from 0.001 to 0.1.
54. The apparatus of claim 49, wherein said biological reagent comprises an enzyme, and substrates of said enzyme present at a concentration of the Michaelis constant (Km) of said enzyme for said substrates.
55. The apparatus of claim 54, wherein said concentration is higher than 1 micromolar.
56. The apparatus of claim 54, wherein said concentration is higher than 50 micromolar.
57. The apparatus of claim 54, wherein said concentration is higher than 100 micromolar.
58. The apparatus of claim 54, wherein said substrates are labeled.
59. The apparatus of claim 54, wherein said individual molecules are products of a reaction between said enzyme and said substrates contained in said biological reagent.
60. The apparatus of claim 49, wherein said individual molecules are DNA/polymerase complexes.
61. The apparatus of claim 49, wherein said individual molecules are biomolecules selected from the group consisting of nucleic acids, polypeptides, and combinations thereof.
62. The apparatus of claim 49, wherein said individual molecules are organic or inorganic chemical compounds.
63. The apparatus of claim 49, wherein said individual molecules are labeled with a detectable label.
64. The apparatus of claim 63, wherein said detectable label is selected from the group consisting of a fluorescent label, a chemiluminescent label, and a radioactive label.
65. The apparatus of claim 49, wherein said waveguides are made of porous film.
66. The apparatus of claim 49, wherein said waveguides are operatively coupled to an optical transmission element transmitting an incident light beam.
67. The apparatus of claim 66, wherein said optical transmission element comprises an element selected from the group consisting of a microlens, a nanolens, an arrayed waveguide grating, an optical switch, an optical fiber, an optical attenuator, a polarization filter, a wave plate, and a delay line.
68. The apparatus of claim 49, wherein said optical system further comprises a photon detector.
69. The apparatus of claim 68, wherein said photon detector is selected from the group consisting of an optical reader, a high efficiency photon detection system, a photodiode, a camera, a charge coupled device, an electron-multiplying charge coupled device, and an intensified charge coupled device.
70. The apparatus of claim 49 that precludes propagation of a majority of an incident electromagnetic energy having a wavelength greater than a cutoff wavelength.
71. The apparatus of claim 70, wherein an individual waveguide of said array comprises a cladding surrounding a core, and wherein said individual waveguide precludes propagation of more than 90% of the incident electromagnetic energy having a wavelength greater than a cutoff wavelength through said core.
72. The apparatus of claim 70, wherein an individual waveguide of said array comprises a cladding surrounding a core, and wherein said individual waveguide precludes propagation of more than 99% of the incident electromagnetic energy having a wavelength greater than a cutoff wavelength through said core.
73. The apparatus of claim 71, wherein each individual waveguide of said array comprises a cladding surrounding a core, and wherein said individual waveguide precludes propagation of more than 90% of the incident electromagnetic energy having a wavelength greater than a cutoff wavelength through said core.
74. The apparatus of claim 70, wherein said cladding comprises a sheet, and one or more holes extending through said sheet where each hole constitutes a core of an individual waveguide.
75. The apparatus of claim 70, wherein an individual waveguide of said array comprises a cladding surrounding a core that is cylindrical.
76. The apparatus of claim 70, wherein an individual waveguide of said array comprises a cladding surrounding a core that is non-cylindrical.
77. The apparatus of claim 70, wherein said cladding comprises a metal alloy.
78. The apparatus of claim 49, wherein each individual waveguide provides an effective observation volume less than 1000 zeptoliters.
79. The apparatus of claim 49, wherein each individual waveguide provides an effective observation volume less than 80 zeptoliters.
80. The apparatus of claim 49, wherein each individual waveguide provides an effective observation volume less than 10 zeptoliters.
81. A kit comprising an array of waveguides of claim 49 and an instruction manual for use of said array.
82. A method of performing multiple chemical reactions involving a plurality of reaction samples, comprising:
(a) providing an array of waveguides of claim 49;
(b) placing the plurality of reaction samples comprising labeled reactants into the waveguides in the array, wherein a separate reaction sample is placed into a different waveguide in the array;
(c) subjecting the array to conditions suitable for formation of products of the chemical reactions;
and (d) detecting the formation of the products with said optical system.
83. The method of claim 82, wherein the step of detecting comprises illuminating the different waveguides with an incident light beam and detecting an optical signal emitted from the reaction samples.
84. The method of claim 82, wherein the chemical reactions involve protein-protein interactions.
85. The method of claim 82, wherein the chemical reactions involve nucleic acid-protein interactions.
86. The method of claim 82, wherein the chemical reactions involve nucleic acid-nucleic acid interactions.
87. A method of detecting a biological analyte, comprising:
optically capturing the analyte within an optical confinement that is created by (a) providing an array of waveguides having a fill fraction greater than 0.0001; and (b) illuminating at least one waveguide within the array that is suspected to contain the analyte with an incident light beam thereby detecting the analyte.
88. The method of claim of claim 87, said array of waveguides having a fill fraction from 0.001 to 0.1.
89. The method of claim 87, wherein said fill fraction is greater than 0.1.
90. The method of claim 87, wherein the analyte is labeled with a detectable label.
91. The method of claim 87, wherein the analyte is selected from the group consisting of nucleic acid, polypeptide, organic compound, and inorganic compound.
92. A method of sequencing a plurality of target nucleic acid molecules, comprising:
(a) providing an array of waveguides having a fill fraction greater than 0.0001;
(b) mixing in the waveguides the plurality of target nucleic acid molecules, primers complementary to the target nucleic acid molecules, polymerization enzymes, and more than one type of nucleotides or nucleotide analogs to be incorporated into a plurality of nascent nucleotide strands, each strand being complementary to a respective target nucleic acid molecule;
(c) subjecting the mixture of step (b) to a polymerization reaction under conditions suitable for formation of the nascent nucleotide strands by template-directed polymerization of the nucleotides or nucleotide analogs;
(d) illuminating the waveguides with an incident light beam; and (e) identifying the nucleotides or the nucleotide analogs incorporated into the each nascent nucleotide strand.
93. The method of claim 92, wherein the step of identifying yields a time sequence of incorporation of the nucleotides provided in step (b).
94. The method of claim 92, wherein the identifying step is effected while the template-directed polymerization is taking place.
95. The method of claim 92, wherein the target nucleic acid molecules are selected from the group consisting of circular DNA molecules, linear DNA molecules, RNA
molecules, and DNA/RNA hybrids.
96. A nucleic acid sequencing method comprising:
subjecting a target nucleic acid molecule to a template-directed polymerization reaction to yield a nascent nucleic acid strand that is complementary to the target nucleic acid molecule in the presence of a plurality of types of nucleotides or nucleotide analogs, and a polymerization enzyme exhibiting strand-displacement activity, wherein said template-directed polymerization reaction processes an identical sequence of nucleotides in the target nucleic acid molecule more than once; registering a time sequence of incorporation of nucleotides or nucleotide analogs into the nascent nucleotide strand; and determining a sequence of nucleotides in the target sequence.
97. The method of claim 96, wherein the target nucleic acid molecule is a circular nucleic acid.
98. The method of claim 97, wherein the target nucleic acid molecule is a circular DNA.
99. The method of claim 97, wherein said template-directed polymerization reaction processes a sequence of nucleotides in the circular nucleic acid multiple times.
100. The method of claim 96, wherein the target nucleic acid molecule comprises a sequence of nucleotides that is present multiple times within the target nucleic acid molecule.
101. The method of claim 96, wherein the target nucleic acid comprises a product of a template dependent replication of a circular nucleic acid.
102. The method of claim 96, wherein the polymerization enzyme is a modified or unmodified 4)29 DNA polymerase.
103. The method of claim 96, wherein said target nucleic acid molecule is sequenced more than once by the polymerization enzyme.
104. The method of claim 96, wherein aid target nucleic acid molecule is sequenced more than twice by the polymerization enzyme.
105. The method of claim 96, wherein the nucleotides or nucleotide analogs further comprise a label.
106. The method of claim 105, wherein the label is attached to the nucleotides or nucleotide analogs at a base, sugar moiety, alpha phosphate, or beta phosphate, of said nucleotides or nucleotide analogs.
107. The method of claim 105, wherein the label is attached to the nucleotides or nucleotide analogs at a terminal phosphate.
108. The method of claim 105, wherein the label is on a terminal phosphate of a nucleotide or nucleotide analog, comprising a phosphate moiety selected from the group consisting of monophosphate, diphosphate, triphosphate, tetraphosphate, pentaphosphate, and hexaphosphate.
109. The method of claim 105, wherein each type of the nucleotides or nucleotide analogs has a different label which is distinguished from one another during said registering step.
110. The method of claim 105, wherein the label is a fluorescence resonance energy transfer donor or acceptor.
111. The method of claim 96, wherein said target nucleic acid molecule is attached to a support.
112. The method of claim 96, wherein said target nucleic acid molecule is hybridized to primer which is attached to a support.
113. The method of claim 96, wherein said polymerization enzyme is attached to a support.
114. The method of claim 96, wherein said registering is performed while the template-directed polymerization reaction is taking place.
115. The method of claim 96, wherein said registering step comprising identifying nucleotides or nucleotide analogs provided in said plurality of types of nucleotides or nucleotide analogs.
116. The method of claim 96, wherein the template-directed polymerization reaction takes place in an optical confinement.
117. The method of claim 96, wherein the template-directed polymerization reaction takes place in an array of optical confinements.
118. The method of claim 96, wherein the template-directed polymerization reaction takes place in a zero-mode waveguide.
119. The method of claim 96, wherein the target nucleic acid molecule and the polymerization enzyme form a single complex immobilized in an optical confinement.
120. The method of claim 119, wherein the array of optical confinements has a fill fraction greater than 0.0001.
121. The method of claim 119, wherein the array of optical confinements has a fill fraction greater than 0.01.
122. The method of claim 119, wherein the array of optical confinements has a fill fraction between 0.00 and 0.1.
123. The method of claim 119, wherein the array of optical confinements has a fill fraction greater than 0.1.
124. The method of claim 117, wherein the array of optical confinements has a density on a substrate exceeding 4x104 confinements per mm2.
125. The method of claim 117, wherein the array of optical confinements has a density on a substrate exceeding 105 confinements per mm2.
126. A method of determining a nucleic acid sequence of a sequence segment of interest, comprising:
providing an individual concatemer molecule comprising a plurality of copies of the sequence segment of interest;
determining nucleotide sequences of the plurality of copies to generate redundant sequence information for said sequence segment of interest, wherein the determining comprises detecting a plurality of luminescent signals, each of which identifies a base in the sequence segment of interest, and further wherein the luminescent signals are indicative of interactions between the individual concatemer molecule and labeled reaction components emitting the luminescent signals; and analyzing the redundant sequence information to determine a nucleic acid sequence of the sequence segment of interest.
127. The method of claim 126, wherein the individual concatemer molecule is a linear template.
128. The method of claim 126, further comprising amplifying a circular template bearing the sequence segment of interest to generate the individual concatemer molecule.
129. The method of claim 126, wherein the plurality of copies of the sequence segment of interest are tandem copies.
130. The method of claim 126, wherein the individual concatemer molecule is immobilized upon a substrate.
131. The method of claim 130, wherein the individual concatemer molecule is covalently immobilized upon the substrate.
132. The method of claim 130, wherein the individual concatemer molecule is noncovalently immobilized upon the substrate.
133. The method of claim 130, wherein the individual concatemer molecule is immobilized upon the substrate via interaction with at least one of the group consisting of a streptavidin/biotin linkage, an avidin/biotin linkage, a carbamate linkage, an ester linkage, an amide, a thiolester, an (N)-functionalized thiourea, a functionalized maleimide, an amino, a disulfide, an amide, a hydrazone linkage, an antibody, a silyl moiety, and an enzyme bound to the substrate.
134. The method of claim 133, wherein the enzyme is a polymerase enzyme.
135. The method of claim 130, wherein a plurality of different individual concatemer molecules are immobilized upon the substrate in a manner that permits optical resolution of the plurality of different individual concatemer molecules from one another.
136. The method of claim 130, wherein the individual concatemer molecule is immobilized in an ordered array.
137. The method of claim 136, wherein the ordered array is an ordered array of optical confinements.
138. The method of claim 137, wherein the optical confinements are zero mode waveguides.
139. The method of claim 126, wherein the luminescent signals are fluorescent signals.
140. The method of claim 126, wherein the detecting is performed using an optical system comprising an excitation source and a photon detector.
141. The method of claim 140, wherein the photon detector is selected from the group consisting of a camera, a fluorescence microscope, a photodiode, an optical reader, a charge-coupled device, and electron-multiplying charge-coupled device, and an intensified charge-coupled device.
142. The method of claim 126, wherein the labeled reaction components are labeled nucleic acids.
143. The method of claim 142, wherein the labeled nucleic acids are labeled nucleotides or nucleotide analogs.
144. The method of claim 126, wherein the determining nucleotide sequences of the plurality of copies comprises a template-directed polymerization reaction.
145. A method for generating redundant sequence information from a single molecule, comprising:
providing a linear template strand comprising multiple repeated copies of a nucleic acid sequence; and determining a nucleotide sequence of the multiple repeated copies in the linear template strand, thereby generating redundant sequence information from a single molecule, wherein the determining comprises detecting a plurality of luminescent signals, each of which identifies a base in the sequence segment of interest, and further wherein the luminescent signals are indicative of interactions between the individual concatemer molecule and labeled reaction components emitting the luminescent signals.
146. The method of claim 145, wherein the nucleic acid sequence is derived from a circular nucleic acid sequence, and wherein the method further comprises synthesizing the linear template strand from the circular nucleic acid sequence such that the linear template strand comprises multiple repeated copies of the circular nucleic acid sequence.
147. The method of claim 146, wherein said synthesizing is performed using a single-molecule amplification strategy.
148. The method of claim 145, wherein the determining comprises immobilization of the linear template strand upon a substrate.
149. The method of claim 148, wherein a plurality of different linear template strands are immobilized upon the substrate in a manner that permits optical resolution of the plurality of different linear template strands from one another.
150. The method of claim 148, wherein the linear template strand is immobilized in an ordered array.
151. The method of claim 145, wherein the detecting is performed using an optical system comprising an excitation source and a photon detector.
152. The method of claim 145, wherein the determining comprises a template-directed polymerization reaction.
153. A method of performing analyses on a plurality of concatemeric nucleic acid templates, comprising:
providing a plurality of concatemeric nucleic acid templates;
providing a substrate;

immobilizing each of the concatemeric nucleic acid templates at a different position on the substrate;
subjecting the substrate to conditions suitable for formation of products of the analyses; and detecting the formation of the products with an optical system, wherein the detecting comprises detecting a plurality of luminescent signals emitted from each of said products, and wherein each of the luminescent signals identifies a base in one of the concatemeric nucleic acid templates, and further wherein the luminescent signals are indicative of interactions between the concatemeric nucleic acid templates and labeled reaction components emitting the luminescent signals, thereby performing analyses on said plurality of concatemeric nucleic acid templates.
154. The method of claim 153, wherein the detecting further comprises illuminating each of said products.
155. The method of claim 153, wherein the luminescent signals emitted from each of said products are optically resolvable from each other.
156. The method of claim 153, wherein the optical system generates and transmits an incident wavelength to the sequencing reactions, and collects and analyzes the luminescent signals emitted from the products.
157. The method of claim 153, wherein the optical system comprises an excitation source and a photon detector.
158. The method of claim 153, wherein the substrate is an ordered array.
159. The method of claim 158, wherein the ordered array comprises a plurality of rows of confinements on a surface of the substrate.
160. The method of claim 159, wherein each of the confinements contains no more than a single of the concatemeric nucleic acid templates.
161. A substrate comprising:
an array of a plurality of optical confinements, wherein each of the plurality of optical confinements comprises a core disposed within a cladding layer that is configured to preclude propagation of electromagnetic radiation through the core, and wherein each core is structured to receive reactants; and wherein the plurality of optical confinements having a density of greater than 4x10 4 confinements per mm2 are aligned upon the substrate in at least a first row or a first column, wherein said first row or said first column is aligned along at least a first axis of the substrate.
162. The substrate of claim 161, wherein the plurality of optical confinements are present on the substrate at a density of at least 8x10 4 confinements per mm2.
163. The substrate of claim 161, wherein the plurality of optical confinements are present on the substrate at a density of at least 1.2x10 5 confinements per mm2.
164. The substrate of claim 161, wherein the array of optical confinements comprises at least 2 rows of optical confinements.
165. The substrate of claim 161, wherein the array of optical confinements comprises at least rows of optical confinements.
166. The substrate of claim 161, wherein the array of optical confinements comprises at least 100 rows of optical confinements.
167. The substrate of claim 164, wherein the at least two rows of optical confinements are further aligned along at least a second axis.
168. The substrate of claim 164, wherein the at least two rows of optical confinements are further aligned in columns.
169. The substrate of claim 164, wherein the at least two rows of optical confinements are further aligned in a hexagonal array.
170. The substrate of claim 161, wherein the optical confinements each comprise a core disposed through a cladding layer that is disposed upon a supporting substrate.
171. The substrate of claim 170, wherein the core comprises a hole disposed through the cladding layer.
172. The substrate of claim 170, wherein the cladding layer comprises a metal layer.
173. The substrate of claim 170, wherein the cladding layer comprises one or more material selected from aluminum, antimony, arsenic, beryllium, bismuth, boron, cadmium, calcium, carbon, cerium, chromium, cobalt, copper, gallium, indium, iron, lead, lithium, magnesium, manganese, mercury, molybdenum, nickel, niobium, phosphorus, silicon, silicate, silicon nitride, gallium phosphide, gallium arsenide, vanadium, and zinc.
174. The substrate of claim 172, wherein the metal layer comprises one or more metal selected from aluminum and chromium.
175. An optical system, comprising:
a substrate comprising:
an array of a plurality of optical confinements, wherein each of the plurality of optical confinements comprises a core disposed within a cladding layer that is configured to preclude propagation of electromagnetic radiation through the core, and wherein each core is structured to receive reactants; and wherein the plurality of optical confinements are aligned upon the substrate in at least a first row or column that is aligned along at least a first axis of the substrate;
an excitation light source positioned to direct excitation light at the array of optical confinements; and a charge coupled device (CCD) positioned to detect emitted signals from the array of optical confinements.
176. The optical system of claim 175, wherein the charge coupled device is selected from an electron multiplying charge coupled device and an intensified charge coupled device (ICCD).
177. The optical system of claim 175, further comprising an optical transmission element for directing excitation light at the array of optical confinements and directing emitted light from the array of optical confinements to the CCD.
178. The optical system of claim 175, wherein the array of confinements and the CCD are aligned so that an optical signal from each confinement is directed to a separate photon detector on the CCD.
179. The optical system of claim 177, wherein the CCD is an electron multiplying CCD
(EMCCD).
180. The optical system of claim 177, wherein the CCD is an intensified CCD
(ICCD).
181. An array of optical confinements for characterizing molecules and/or monitoring a chemical reaction, said array having a surface density exceeding 4x10 4 confinements per mm2, wherein individual confinement in the array permits resolution of individual molecules present at a concentration higher than about 1 micromolar.
182. An array of optical confinements for characterizing molecules and/or monitoring a chemical reaction, said array having a surface density exceeding 4x10 4 confinements per mm2, wherein individual confinement in the array provides an effective observation volume less than about 1000 zeptoliters.
183. A method of detecting interactions among a plurality of molecules, comprising:
placing the plurality of molecules in close proximity to an array of zero-mode waveguides, wherein individual waveguides in the array are separated by a distance sufficient to yield detectable intensities of diffractive scattering at multiple diffracted orders upon illuminating the array with an incident wavelength;
illuminating the array of zero-mode waveguides with an incident wavelength;
and detecting a change in the intensities of diffractive scattering of the incident wavelength at the multiple diffracted orders, thereby detecting the interactions among a plurality of molecules.
184. A method of reducing diffractive scattering upon illuminating an array of optical confinement with an incident wavelength, wherein the array comprises at least a first optical confinement and a second confinement, said method comprising:
forming the array of optical confinements wherein the first zero-mode waveguide is separated from the second zero-mode waveguide by a distance such that upon illumination with the incident wavelength, intensity of diffractive scattering resulting from the first zero-mode waveguide at a given angle is less than that if the first zero-mode waveguide were illuminated with the same incident wavelength in the absence of the second zero-mode waveguide.
185. A method of fabricating an array of optical confinements that exhibits a minimal intensity of diffractive scattering of an incident wavelength, comprising:
providing a substrate; and forming the array of optical confinements on the substrate such that individual confinements in the array are separated from each other at a distance less than one half of the wavelength.
186. A method of creating a plurality of optical confinements having a surface density exceeding 4x10 4 confinements per mm2, wherein individual confinement in the array permits resolution of individual molecules present at a concentration higher than at least about 1 micromolar, comprising:
providing a substrate;
forming an array of optical confinements having a surface density exceeding 4x10 4 confinements per mm2, wherein the individual confinement comprises a zero-mode waveguide comprising: a cladding surrounding a core, wherein said cladding is configured to preclude propagation of electromagnetic energy of a frequency less than a cutoff frequency longitudinally through the core of the zero-mode waveguide; and illuminating the array with an electromagnetic radiation of a frequency less than the cutoff frequency, thereby creating a plurality of optical confinements.
CA2579150A 2004-09-17 2005-09-16 Apparatus and method for analysis of molecules Active CA2579150C (en)

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
US10/944,106 2004-09-17
US10/944,106 US7170050B2 (en) 2004-09-17 2004-09-17 Apparatus and methods for optical analysis of molecules
US64900905P 2005-01-31 2005-01-31
US60/649,009 2005-01-31
US65184605P 2005-02-09 2005-02-09
US60/651,846 2005-02-09
PCT/US2005/033052 WO2006044078A2 (en) 2004-09-17 2005-09-16 Apparatus and method for analysis of molecules

Publications (2)

Publication Number Publication Date
CA2579150A1 CA2579150A1 (en) 2006-04-27
CA2579150C true CA2579150C (en) 2014-11-25

Family

ID=36073574

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2579150A Active CA2579150C (en) 2004-09-17 2005-09-16 Apparatus and method for analysis of molecules

Country Status (8)

Country Link
US (4) US7476503B2 (en)
EP (2) EP3415641B1 (en)
JP (3) JP2008513782A (en)
CN (1) CN101914620B (en)
AU (1) AU2005296200B2 (en)
CA (1) CA2579150C (en)
GB (1) GB2423819B (en)
WO (1) WO2006044078A2 (en)

Families Citing this family (638)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070172866A1 (en) * 2000-07-07 2007-07-26 Susan Hardin Methods for sequence determination using depolymerizing agent
CN101525660A (en) * 2000-07-07 2009-09-09 维西根生物技术公司 An instant sequencing methodology
WO2002044425A2 (en) * 2000-12-01 2002-06-06 Visigen Biotechnologies, Inc. Enzymatic nucleic acid synthesis: compositions and methods for altering monomer incorporation fidelity
US7668697B2 (en) * 2006-02-06 2010-02-23 Andrei Volkov Method for analyzing dynamic detectable events at the single molecule level
US20040067532A1 (en) 2002-08-12 2004-04-08 Genetastix Corporation High throughput generation and affinity maturation of humanized antibody
US20090186343A1 (en) * 2003-01-28 2009-07-23 Visigen Biotechnologies, Inc. Methods for preparing modified biomolecules, modified biomolecules and methods for using same
US7476503B2 (en) * 2004-09-17 2009-01-13 Pacific Biosciences Of California, Inc. Apparatus and method for performing nucleic acid analysis
US7170050B2 (en) * 2004-09-17 2007-01-30 Pacific Biosciences Of California, Inc. Apparatus and methods for optical analysis of molecules
US8535616B2 (en) * 2005-08-02 2013-09-17 Moxtek, Inc. Sub-wavelength metallic apertures as light enhancement devices
US7763423B2 (en) * 2005-09-30 2010-07-27 Pacific Biosciences Of California, Inc. Substrates having low density reactive groups for monitoring enzyme activity
WO2007041621A2 (en) * 2005-10-03 2007-04-12 Xingsheng Sean Ling Hybridization assisted nanopore sequencing
JP2009519717A (en) * 2005-12-16 2009-05-21 アプレラ コーポレイション Method and system for fixed phase sequencing
WO2007091280A1 (en) * 2006-02-06 2007-08-16 Stmicroelectronics S.R.L. Optical apparatus and method for the inspection of nucleic acid probes by polarized radiation
US7692783B2 (en) * 2006-02-13 2010-04-06 Pacific Biosciences Of California Methods and systems for simultaneous real-time monitoring of optical signals from multiple sources
US7715001B2 (en) 2006-02-13 2010-05-11 Pacific Biosciences Of California, Inc. Methods and systems for simultaneous real-time monitoring of optical signals from multiple sources
US7995202B2 (en) 2006-02-13 2011-08-09 Pacific Biosciences Of California, Inc. Methods and systems for simultaneous real-time monitoring of optical signals from multiple sources
US9528939B2 (en) 2006-03-10 2016-12-27 Indx Lifecare, Inc. Waveguide-based optical scanning systems
US9423397B2 (en) 2006-03-10 2016-08-23 Indx Lifecare, Inc. Waveguide-based detection system with scanning light source
US7951583B2 (en) 2006-03-10 2011-05-31 Plc Diagnostics, Inc. Optical scanning system
US8288157B2 (en) 2007-09-12 2012-10-16 Plc Diagnostics, Inc. Waveguide-based optical scanning systems
US9976192B2 (en) 2006-03-10 2018-05-22 Ldip, Llc Waveguide-based detection system with scanning light source
US8975216B2 (en) 2006-03-30 2015-03-10 Pacific Biosciences Of California Articles having localized molecules disposed thereon and methods of producing same
CN101467082B (en) * 2006-06-12 2011-12-14 加利福尼亚太平洋生物科学公司 Substrates for performing analytical reactions
EP2029780A4 (en) * 2006-06-16 2010-03-31 Pacific Biosciences California Controlled initiation of primer extension
US20080241951A1 (en) * 2006-07-20 2008-10-02 Visigen Biotechnologies, Inc. Method and apparatus for moving stage detection of single molecular events
US20080241938A1 (en) * 2006-07-20 2008-10-02 Visigen Biotechnologies, Inc. Automated synthesis or sequencing apparatus and method for making and using same
US20080091005A1 (en) * 2006-07-20 2008-04-17 Visigen Biotechnologies, Inc. Modified nucleotides, methods for making and using same
EP4220138A1 (en) * 2006-09-01 2023-08-02 Pacific Biosciences of California, Inc. Substrates, systems and methods for analyzing materials
US8207509B2 (en) 2006-09-01 2012-06-26 Pacific Biosciences Of California, Inc. Substrates, systems and methods for analyzing materials
US9096953B2 (en) * 2006-09-29 2015-08-04 Intel Corporation Method for high throughput, high volume manufacturing of biomolecule micro arrays
US11747279B2 (en) * 2006-12-06 2023-09-05 Mohammad A. Mazed Optical biomodule for detection of diseases at an early onset
US10529003B2 (en) * 2008-04-07 2020-01-07 Mohammad A. Mazed Optical biomodule for detection of diseases at an early onset
KR20150075119A (en) * 2006-12-28 2015-07-02 생-고뱅 세라믹스 앤드 플라스틱스, 인코포레이티드 A sapphire substrate
CN101616772B (en) * 2006-12-28 2012-03-21 圣戈本陶瓷及塑料股份有限公司 Sapphire substrates and methods of making same
RU2414550C1 (en) * 2006-12-28 2011-03-20 Сэнт-Гобэн Керамикс Энд Пластикс, Инк. Sapphire substrate (versions)
US8740670B2 (en) 2006-12-28 2014-06-03 Saint-Gobain Ceramics & Plastics, Inc. Sapphire substrates and methods of making same
US11940413B2 (en) 2007-02-05 2024-03-26 IsoPlexis Corporation Methods and devices for sequencing nucleic acids in smaller batches
US8551704B2 (en) * 2007-02-16 2013-10-08 Pacific Biosciences Of California, Inc. Controllable strand scission of mini circle DNA
DE102007016699A1 (en) * 2007-04-04 2008-10-09 Synentec Gmbh Biochip for the fluorescence analysis of individual transporters
US9121843B2 (en) 2007-05-08 2015-09-01 Trustees Of Boston University Chemical functionalization of solid-state nanopores and nanopore arrays and applications thereof
US7714303B2 (en) * 2007-05-10 2010-05-11 Pacific Biosciences Of California, Inc. Methods and systems for analyzing fluorescent materials with reduced authofluorescence
US20100167413A1 (en) * 2007-05-10 2010-07-01 Paul Lundquist Methods and systems for analyzing fluorescent materials with reduced autofluorescence
AU2008251861B2 (en) * 2007-05-10 2014-03-20 Pacific Biosciences Of California, Inc. Methods and systems for analyzing fluorescent materials with reduced autofluorescence
US20080277595A1 (en) * 2007-05-10 2008-11-13 Pacific Biosciences Of California, Inc. Highly multiplexed confocal detection systems and methods of using same
AU2008261935B2 (en) * 2007-06-06 2013-05-02 Pacific Biosciences Of California, Inc. Methods and processes for calling bases in sequence by incorporation methods
US8703422B2 (en) 2007-06-06 2014-04-22 Pacific Biosciences Of California, Inc. Methods and processes for calling bases in sequence by incorporation methods
US9151751B2 (en) * 2007-06-29 2015-10-06 Applied Biosystems, Llc Systems and methods for electronic detection with nanoFETS
AU2008282862B2 (en) * 2007-07-26 2014-07-31 Pacific Biosciences Of California, Inc. Molecular redundant sequencing
WO2009023489A1 (en) * 2007-08-14 2009-02-19 Massachusetts Institute Of Technology Nanoscale imaging via absorption modulation
US8877688B2 (en) 2007-09-14 2014-11-04 Adimab, Llc Rationally designed, synthetic antibody libraries and uses therefor
BRPI0816785A2 (en) 2007-09-14 2017-05-02 Adimab Inc rationally designed synthetic antibody libraries, and uses thereof
US20090118129A1 (en) * 2007-09-28 2009-05-07 Pacific Biosciences Of California, Inc. Virtual reads for readlength enhancement
US7960116B2 (en) 2007-09-28 2011-06-14 Pacific Biosciences Of California, Inc. Nucleic acid sequencing methods and systems
US8003330B2 (en) * 2007-09-28 2011-08-23 Pacific Biosciences Of California, Inc. Error-free amplification of DNA for clonal sequencing
EP2201136B1 (en) 2007-10-01 2017-12-06 Nabsys 2.0 LLC Nanopore sequencing by hybridization of probes to form ternary complexes and variable range alignment
WO2009073201A2 (en) 2007-12-04 2009-06-11 Pacific Biosciences Of California, Inc. Alternate labeling strategies for single molecule sequencing
EP2245444A4 (en) * 2008-01-10 2014-04-16 Pacific Biosciences California Methods and systems for analysis of fluorescent reactions with modulated excitation
US8652781B2 (en) 2008-02-12 2014-02-18 Pacific Biosciences Of California, Inc. Cognate sampling kinetics
US8236499B2 (en) 2008-03-28 2012-08-07 Pacific Biosciences Of California, Inc. Methods and compositions for nucleic acid sample preparation
MX2010010600A (en) 2008-03-28 2011-03-30 Pacific Biosciences California Inc Compositions and methods for nucleic acid sequencing.
US8628940B2 (en) * 2008-09-24 2014-01-14 Pacific Biosciences Of California, Inc. Intermittent detection during analytical reactions
US8420366B2 (en) * 2008-03-31 2013-04-16 Pacific Biosciences Of California, Inc. Generation of modified polymerases for improved accuracy in single molecule sequencing
US8999676B2 (en) 2008-03-31 2015-04-07 Pacific Biosciences Of California, Inc. Recombinant polymerases for improved single molecule sequencing
WO2009145818A1 (en) 2008-03-31 2009-12-03 Pacific Biosciences Of California, Inc Single molecule loading methods and compositions
EP2274446B1 (en) * 2008-03-31 2015-09-09 Pacific Biosciences of California, Inc. Two slow-step polymerase enzyme systems and methods
EP2942404B1 (en) * 2008-03-31 2016-11-23 Pacific Biosciences of California, Inc. Generation of modified polymerases for improved accuracy in single molecule sequencing
US20090247426A1 (en) * 2008-03-31 2009-10-01 Pacific Biosciences Of California, Inc. Focused library generation
EP2274445A2 (en) 2008-04-11 2011-01-19 University of Utah Research Foundation Methods and compositions related to quantitative array based methylation analysis
US20110250402A1 (en) * 2008-06-02 2011-10-13 Applied Biosystems, Llc Localization of near-field resonances in bowtie antennae: influence of adhesion layers
GB2461026B (en) 2008-06-16 2011-03-09 Plc Diagnostics Inc System and method for nucleic acids sequencing by phased synthesis
US8198023B2 (en) 2008-08-05 2012-06-12 Pacific Biosciences Of California, Inc. Prevention and alleviation of steric hindrance during single molecule nucleic acid synthesis by a polymerase
US20100227327A1 (en) * 2008-08-08 2010-09-09 Xiaoliang Sunney Xie Methods and compositions for continuous single-molecule nucleic acid sequencing by synthesis with fluorogenic nucleotides
US20100036110A1 (en) * 2008-08-08 2010-02-11 Xiaoliang Sunney Xie Methods and compositions for continuous single-molecule nucleic acid sequencing by synthesis with fluorogenic nucleotides
EP2342362B1 (en) * 2008-09-03 2017-03-01 Nabsys 2.0 LLC Use of longitudinally displaced nanoscale electrodes for voltage sensing of biomolecules and other analytes in fluidic channels
US9650668B2 (en) 2008-09-03 2017-05-16 Nabsys 2.0 Llc Use of longitudinally displaced nanoscale electrodes for voltage sensing of biomolecules and other analytes in fluidic channels
US8262879B2 (en) 2008-09-03 2012-09-11 Nabsys, Inc. Devices and methods for determining the length of biopolymers and distances between probes bound thereto
US8795961B2 (en) * 2008-09-05 2014-08-05 Pacific Biosciences Of California, Inc. Preparations, compositions, and methods for nucleic acid sequencing
CA2735979A1 (en) * 2008-09-05 2010-03-11 Pacific Biosciences Of California, Inc. Engineering polymerases and reaction conditions for modified incorporation properties
US8383345B2 (en) 2008-09-12 2013-02-26 University Of Washington Sequence tag directed subassembly of short sequencing reads into long sequencing reads
CA2974241C (en) 2008-09-16 2020-01-07 Pacific Biosciences Of California, Inc. Substrates and optical systems and methods of use thereof
US8921046B2 (en) * 2008-09-19 2014-12-30 Pacific Biosciences Of California, Inc. Nucleic acid sequence analysis
US8481264B2 (en) * 2008-09-19 2013-07-09 Pacific Biosciences Of California, Inc. Immobilized nucleic acid complexes for sequence analysis
WO2010036287A1 (en) * 2008-09-24 2010-04-01 Pacific Biosciences Of California, Inc. Intermittent detection during analytical reactions
US8383369B2 (en) * 2008-09-24 2013-02-26 Pacific Biosciences Of California, Inc. Intermittent detection during analytical reactions
EP2362947B1 (en) 2008-09-30 2017-05-10 Pacific Biosciences of California, Inc. Ultra-high multiplex analytical systems and methods
AU2009319907B2 (en) 2008-11-03 2015-10-01 The Regents Of The University Of California Methods for detecting modification resistant nucleic acids
US8486630B2 (en) 2008-11-07 2013-07-16 Industrial Technology Research Institute Methods for accurate sequence data and modified base position determination
US8993230B2 (en) * 2008-12-04 2015-03-31 Pacific Biosciences of Californ, Inc. Asynchronous sequencing of biological polymers
US9175338B2 (en) 2008-12-11 2015-11-03 Pacific Biosciences Of California, Inc. Methods for identifying nucleic acid modifications
CN102317473A (en) * 2008-12-11 2012-01-11 加利福尼亚太平洋生物科学股份有限公司 Shenzhen tcl new technology co. , ltd
US20230148447A9 (en) 2008-12-11 2023-05-11 Pacific Biosciences Of California, Inc. Classification of nucleic acid templates
WO2010075188A2 (en) 2008-12-23 2010-07-01 Illumina Inc. Multibase delivery for long reads in sequencing by synthesis protocols
KR20110138340A (en) * 2009-01-20 2011-12-27 더 보드 어브 트러스티스 어브 더 리랜드 스탠포드 주니어 유니버시티 Single cell gene expression for diagnosis, prognosis and identification of drug targets
US9005548B2 (en) * 2009-02-25 2015-04-14 California Institute Of Technology Methods for fabricating high aspect ratio probes and deforming high aspect ratio nanopillars and micropillars
US9778188B2 (en) * 2009-03-11 2017-10-03 Industrial Technology Research Institute Apparatus and method for detection and discrimination molecular object
US8455260B2 (en) * 2009-03-27 2013-06-04 Massachusetts Institute Of Technology Tagged-fragment map assembly
EP2411536B1 (en) * 2009-03-27 2014-09-17 Nabsys, Inc. Methods for analyzing biomolecules and probes bound thereto
US20100261185A1 (en) 2009-03-27 2010-10-14 Life Technologies Corporation Labeled enzyme compositions, methods and systems
WO2010117420A2 (en) 2009-03-30 2010-10-14 Pacific Biosciences Of California, Inc. Fret-labeled compounds and uses therefor
US8986928B2 (en) 2009-04-10 2015-03-24 Pacific Biosciences Of California, Inc. Nanopore sequencing devices and methods
EP2425023B1 (en) 2009-04-27 2015-12-23 Pacific Biosciences of California, Inc. Real-time sequencing methods and systems
EP2425286B1 (en) * 2009-04-29 2020-06-24 Ldip, Llc Waveguide-based detection system with scanning light source
EP2427572B1 (en) * 2009-05-01 2013-08-28 Illumina, Inc. Sequencing methods
US20120064527A1 (en) * 2009-05-27 2012-03-15 Akira Maekawa Nucleic acid analysis device, nucleic acid analysis apparatus, and nucleic acid analysis method
US8246799B2 (en) * 2009-05-28 2012-08-21 Nabsys, Inc. Devices and methods for analyzing biomolecules and probes bound thereto
US8316738B2 (en) * 2009-06-10 2012-11-27 Magna Powertrain Of America, Inc. Compact transfer case with beveloid gearset
US9063156B2 (en) 2009-06-12 2015-06-23 Pacific Biosciences Of California, Inc. Real-time analytical methods and systems
US8609421B2 (en) * 2009-06-12 2013-12-17 Pacific Biosciences Of California, Inc. Single-molecule real-time analysis of protein synthesis
EP2446467A4 (en) 2009-06-26 2014-07-02 California Inst Of Techn Methods for fabricating passivated silicon nanowires and devices thus obtained
US9671558B2 (en) * 2009-06-30 2017-06-06 Intel Corporation Chemically induced optical signals and DNA sequencing
US8501406B1 (en) 2009-07-14 2013-08-06 Pacific Biosciences Of California, Inc. Selectively functionalized arrays
WO2011014811A1 (en) 2009-07-31 2011-02-03 Ibis Biosciences, Inc. Capture primers and capture sequence linked solid supports for molecular diagnostic tests
WO2011017656A2 (en) * 2009-08-06 2011-02-10 Ibis Biosciences, Inc. Non-mass determined base compositions for nucleic acid detection
EP2816111B1 (en) 2009-08-14 2016-04-13 Epicentre Technologies Corporation Methods, compositions, and kits for generating rRNA-depleted samples or isolating rRNA from samples
US8367159B2 (en) 2009-09-11 2013-02-05 Pacific Biosciences Of California, Inc. Methods for producing ZMWs with islands of functionality
US8906670B2 (en) 2009-09-11 2014-12-09 Pacific Bioscience Of California, Inc. Zero-mode waveguides with non-reflecting walls
AU2010301128B2 (en) 2009-09-30 2014-09-18 Quantapore, Inc. Ultrafast sequencing of biological polymers using a labeled nanopore
WO2011050127A1 (en) * 2009-10-22 2011-04-28 The University Of North Carolina At Chapel Hill Particles for the effective fabrication and utilization of micro- and nanoarrays
WO2011063163A2 (en) * 2009-11-19 2011-05-26 California Institute Of Technology Methods for fabricating self-aligning arrangements on semiconductors
US9018684B2 (en) 2009-11-23 2015-04-28 California Institute Of Technology Chemical sensing and/or measuring devices and methods
JP5873023B2 (en) 2009-12-01 2016-03-01 オックスフォード ナノポール テクノロジーズ リミテッド Biochemical analyzer
WO2011093939A1 (en) 2010-02-01 2011-08-04 Illumina, Inc. Focusing methods and optical systems and assemblies using the same
US8467061B2 (en) 2010-02-19 2013-06-18 Pacific Biosciences Of California, Inc. Integrated analytical system and method
US8994946B2 (en) 2010-02-19 2015-03-31 Pacific Biosciences Of California, Inc. Integrated analytical system and method
CN202281746U (en) * 2010-03-06 2012-06-20 伊鲁米那股份有限公司 Measuring equipment for detecting optical signal from sample as well as optical module and optical system for measuring equipment
JP5926693B2 (en) * 2010-03-12 2016-05-25 イョットペーテー・ペプタイド・テクノロジーズ・ゲゼルシャフト・ミット・ベシュレンクテル・ハフツングJpt Peptide Technologies Gmbh Method for determining peptide concentration
US9482615B2 (en) 2010-03-15 2016-11-01 Industrial Technology Research Institute Single-molecule detection system and methods
US9371598B2 (en) 2010-04-05 2016-06-21 Prognosys Biosciences, Inc. Spatially encoded biological assays
US10787701B2 (en) 2010-04-05 2020-09-29 Prognosys Biosciences, Inc. Spatially encoded biological assays
US20190300945A1 (en) 2010-04-05 2019-10-03 Prognosys Biosciences, Inc. Spatially Encoded Biological Assays
US8652779B2 (en) 2010-04-09 2014-02-18 Pacific Biosciences Of California, Inc. Nanopore sequencing using charge blockade labels
TWI534431B (en) 2010-04-28 2016-05-21 加州太平洋生物科學公司 Method for forming nanoscale apertures having islands of functionality
JP2013527769A (en) 2010-05-06 2013-07-04 アイビス バイオサイエンシズ インコーポレイティッド Integrated sample preparation system and stabilized enzyme mixture
WO2011140433A2 (en) 2010-05-07 2011-11-10 The Board Of Trustees Of The Leland Stanford Junior University Measurement and comparison of immune diversity by high-throughput sequencing
US9670243B2 (en) 2010-06-02 2017-06-06 Industrial Technology Research Institute Compositions and methods for sequencing nucleic acids
US8865077B2 (en) 2010-06-11 2014-10-21 Industrial Technology Research Institute Apparatus for single-molecule detection
US8865078B2 (en) 2010-06-11 2014-10-21 Industrial Technology Research Institute Apparatus for single-molecule detection
WO2012009206A2 (en) 2010-07-12 2012-01-19 Pacific Biosciences Of California, Inc. Sequencing reactions with alkali metal cations for pulse width control
US9777320B2 (en) 2010-08-25 2017-10-03 Pacific Biosciences Of California, Inc. Molecular adaptors for dye conjugates
US9938590B2 (en) 2010-09-16 2018-04-10 Gen-Probe Incorporated Capture probes immobilizable via L-nucleotide tail
AU2011301935B2 (en) 2010-09-16 2015-06-11 Ibis Biosciences, Inc. Stabilization of ozone-labile fluorescent dyes by thiourea
US8715933B2 (en) 2010-09-27 2014-05-06 Nabsys, Inc. Assay methods using nicking endonucleases
US8753816B2 (en) 2010-10-26 2014-06-17 Illumina, Inc. Sequencing methods
EP2635703B1 (en) 2010-11-01 2018-03-21 Gen-Probe Incorporated Integrated capture and amplification of target nucleic acid for sequencing
US9074251B2 (en) 2011-02-10 2015-07-07 Illumina, Inc. Linking sequence reads using paired code tags
CA2821299C (en) 2010-11-05 2019-02-12 Frank J. Steemers Linking sequence reads using paired code tags
US8859201B2 (en) 2010-11-16 2014-10-14 Nabsys, Inc. Methods for sequencing a biomolecule by detecting relative positions of hybridized probes
EP2643484A4 (en) 2010-11-22 2014-04-16 Univ California Methods of identifying a cellular nascent rna transcript
WO2012092265A1 (en) 2010-12-27 2012-07-05 Ibis Biosciences, Inc. Nucleic acid sample preparation methods and compositions
US8951781B2 (en) 2011-01-10 2015-02-10 Illumina, Inc. Systems, methods, and apparatuses to image a sample for biological or chemical analysis
EP2670864B1 (en) 2011-01-31 2017-03-08 Illumina, Inc. Methods for reducing nucleic acid damage
WO2012106546A2 (en) 2011-02-02 2012-08-09 University Of Washington Through Its Center For Commercialization Massively parallel continguity mapping
WO2012109574A2 (en) 2011-02-11 2012-08-16 Nabsys, Inc. Assay methods using dna binding proteins
EP2689028B1 (en) 2011-03-23 2017-08-30 Pacific Biosciences Of California, Inc. Isolation of polymerase-nucleic acid complexes and loading onto substrates
US9611510B2 (en) 2011-04-06 2017-04-04 The University Of Chicago Composition and methods related to modification of 5-methylcytosine (5-mC)
GB201106254D0 (en) 2011-04-13 2011-05-25 Frisen Jonas Method and product
EP2718465B1 (en) 2011-06-09 2022-04-13 Illumina, Inc. Method of making an analyte array
US9670538B2 (en) 2011-08-05 2017-06-06 Ibis Biosciences, Inc. Nucleic acid sequencing by electrochemical detection
AU2012304520B2 (en) 2011-09-06 2016-06-16 Gen-Probe Incorporated Circularized templates for sequencing
EP4219741A3 (en) 2011-09-06 2023-08-23 Gen-Probe Incorporated Closed nucleic acid structures
WO2013049135A1 (en) 2011-09-26 2013-04-04 Gen-Probe Incorporated Algorithms for sequence determinations
WO2013056241A2 (en) 2011-10-14 2013-04-18 Pacific Biosciences Of California, Inc. Real-time redox sequencing
CN104080958A (en) 2011-10-19 2014-10-01 纽亘技术公司 Compositions and methods for directional nucleic acid amplification and sequencing
WO2013063308A1 (en) 2011-10-25 2013-05-02 University Of Massachusetts An enzymatic method to enrich for capped rna, kits for performing same, and compositions derived therefrom
US8778849B2 (en) 2011-10-28 2014-07-15 Illumina, Inc. Microarray fabrication system and method
CA2854023A1 (en) 2011-11-07 2013-05-16 Illumina, Inc. Integrated sequencing apparatuses and methods of use
US9200274B2 (en) 2011-12-09 2015-12-01 Illumina, Inc. Expanded radix for polymeric tags
US20140349858A1 (en) 2011-12-22 2014-11-27 Ibis Bioscience, Inc. Amplification of a sequence from a ribonucleic acid
ES2626058T3 (en) 2011-12-22 2017-07-21 Ibis Biosciences, Inc. Primers and amplification methods
US10150993B2 (en) 2011-12-22 2018-12-11 Ibis Biosciences, Inc. Macromolecule positioning by electrical potential
WO2013102081A2 (en) 2011-12-29 2013-07-04 Ibis Biosciences, Inc. Macromolecule delivery to nanowells
CA2863121A1 (en) 2011-12-30 2013-07-04 Abbott Molecular Inc. Microorganism nucleic acid purification from host samples
WO2013106737A1 (en) 2012-01-13 2013-07-18 Data2Bio Genotyping by next-generation sequencing
WO2013105025A1 (en) * 2012-01-13 2013-07-18 Koninklijke Philips N.V. Dna sequencing with reagent recycling on wiregrid
GB2513793B (en) 2012-01-26 2016-11-02 Nugen Tech Inc Compositions and methods for targeted nucleic acid sequence enrichment and high efficiency library generation
US9238836B2 (en) 2012-03-30 2016-01-19 Pacific Biosciences Of California, Inc. Methods and compositions for sequencing modified nucleic acids
EP3222627B1 (en) 2012-02-15 2019-08-07 Pacific Biosciences of California, Inc. Polymerase enzyme substrates with protein shield
NO2694769T3 (en) 2012-03-06 2018-03-03
WO2013138724A2 (en) * 2012-03-16 2013-09-19 Life Technologies Corporation Systems and methods for loading liquid samples
JP2015511015A (en) * 2012-03-16 2015-04-13 ライフ テクノロジーズ コーポレーション System and method for containing biological samples
EP2828218B9 (en) 2012-03-20 2021-04-07 University Of Washington Through Its Center For Commercialization Methods of lowering the error rate of massively parallel dna sequencing using duplex consensus sequencing
US9803239B2 (en) 2012-03-29 2017-10-31 Complete Genomics, Inc. Flow cells for high density array chips
US20130261984A1 (en) 2012-03-30 2013-10-03 Illumina, Inc. Methods and systems for determining fetal chromosomal abnormalities
AU2013240166A1 (en) 2012-03-30 2014-10-30 Pacific Biosciences Of California, Inc. Methods and composition for sequencing modified nucleic acids
JP6159391B2 (en) 2012-04-03 2017-07-05 イラミーナ インコーポレーテッド Integrated read head and fluid cartridge useful for nucleic acid sequencing
US20130274148A1 (en) 2012-04-11 2013-10-17 Illumina, Inc. Portable genetic detection and analysis system and method
WO2013163207A1 (en) 2012-04-24 2013-10-31 Pacific Biosciences Of California, Inc. Identification of 5-methyl-c in nucleic acid templates
US10202642B2 (en) 2012-05-02 2019-02-12 Ibis Biosciences, Inc. DNA sequencing
EP2844774B1 (en) 2012-05-02 2018-07-18 Ibis Biosciences, Inc. Dna sequencing
ES2833524T3 (en) 2012-05-02 2021-06-15 Ibis Biosciences Inc DNA sequencing
US9315864B2 (en) 2012-05-18 2016-04-19 Pacific Biosciences Of California, Inc. Heteroarylcyanine dyes with sulfonic acid substituents
US10458915B2 (en) 2012-05-18 2019-10-29 Pacific Biosciences Of California, Inc. Heteroarylcyanine dyes
US9012022B2 (en) 2012-06-08 2015-04-21 Illumina, Inc. Polymer coatings
WO2013185137A1 (en) 2012-06-08 2013-12-12 Pacific Biosciences Of California, Inc. Modified base detection with nanopore sequencing
US9267168B2 (en) 2012-06-12 2016-02-23 Pacific Biosciences Of California, Inc. Methods and compositions for isolating template nucleic acids
US8895249B2 (en) 2012-06-15 2014-11-25 Illumina, Inc. Kinetic exclusion amplification of nucleic acid libraries
US9372308B1 (en) 2012-06-17 2016-06-21 Pacific Biosciences Of California, Inc. Arrays of integrated analytical devices and methods for production
EP2861787B1 (en) 2012-06-18 2017-09-20 Nugen Technologies, Inc. Compositions and methods for negative selection of non-desired nucleic acid sequences
US20150167084A1 (en) 2012-07-03 2015-06-18 Sloan Kettering Institute For Cancer Research Quantitative Assessment of Human T-Cell Repertoire Recovery After Allogeneic Hematopoietic Stem Cell Transplantation
US20150011396A1 (en) 2012-07-09 2015-01-08 Benjamin G. Schroeder Methods for creating directional bisulfite-converted nucleic acid libraries for next generation sequencing
NL2017959B1 (en) 2016-12-08 2018-06-19 Illumina Inc Cartridge assembly
US10895534B2 (en) 2012-08-20 2021-01-19 Illumina, Inc. Method and system for fluorescence lifetime based sequencing
US9637782B2 (en) 2012-09-28 2017-05-02 Pacific Biosciences Of California, Inc. Charged triplet-state quenchers for mitigation of photo-induced damage
US9399766B2 (en) 2012-10-01 2016-07-26 Pacific Biosciences Of California, Inc. Recombinant polymerases for incorporation of protein shield nucleotide analogs
EP3447150A1 (en) 2012-10-16 2019-02-27 Abbott Molecular Inc. Methods and apparatus to sequence a nucleic acid
US9181583B2 (en) 2012-10-23 2015-11-10 Illumina, Inc. HLA typing using selective amplification and sequencing
US9651539B2 (en) 2012-10-28 2017-05-16 Quantapore, Inc. Reducing background fluorescence in MEMS materials by low energy ion beam treatment
WO2014071070A1 (en) 2012-11-01 2014-05-08 Pacific Biosciences Of California, Inc. Compositions and methods for selection of nucleic acids
US9223084B2 (en) 2012-12-18 2015-12-29 Pacific Biosciences Of California, Inc. Illumination of optical analytical devices
US9914966B1 (en) 2012-12-20 2018-03-13 Nabsys 2.0 Llc Apparatus and methods for analysis of biomolecules using high frequency alternating current excitation
US9683230B2 (en) 2013-01-09 2017-06-20 Illumina Cambridge Limited Sample preparation on a solid support
EP2956550B1 (en) 2013-01-18 2020-04-08 Nabsys 2.0 LLC Enhanced probe binding
US9562269B2 (en) 2013-01-22 2017-02-07 The Board Of Trustees Of The Leland Stanford Junior University Haplotying of HLA loci with ultra-deep shotgun sequencing
US9805407B2 (en) 2013-01-25 2017-10-31 Illumina, Inc. Methods and systems for using a cloud computing environment to configure and sell a biological sample preparation cartridge and share related data
WO2014130900A1 (en) 2013-02-22 2014-08-28 Pacific Biosciences Of California, Inc. Integrated illumination of optical analytical devices
US9512422B2 (en) 2013-02-26 2016-12-06 Illumina, Inc. Gel patterned surfaces
AU2013382089B2 (en) 2013-03-13 2018-05-10 Illumina, Inc. Multilayer fluidic devices and methods for their fabrication
WO2014142850A1 (en) 2013-03-13 2014-09-18 Illumina, Inc. Methods and compositions for nucleic acid sequencing
US10421996B2 (en) 2013-03-14 2019-09-24 Illumina, Inc. Modified polymerases for improved incorporation of nucleotide analogues
US9193998B2 (en) 2013-03-15 2015-11-24 Illumina, Inc. Super resolution imaging
DK2970356T3 (en) 2013-03-15 2018-08-27 Illumina Cambridge Ltd Modified nucleosides or nucleotides
EP2970366B1 (en) 2013-03-15 2019-01-16 Ibis Biosciences, Inc. Nucleotide analogs for sequencing
US20140274747A1 (en) 2013-03-15 2014-09-18 Illumina, Inc. Super resolution imaging
WO2014144092A1 (en) 2013-03-15 2014-09-18 Nugen Technologies, Inc. Sequential sequencing
SG10201708498VA (en) 2013-04-17 2017-11-29 Agency Science Tech & Res Method for generating extended sequence reads
CN111187811B (en) 2013-05-06 2024-03-08 加利福尼亚太平洋生物科学股份有限公司 Real-time electronic sequencing
US9862997B2 (en) 2013-05-24 2018-01-09 Quantapore, Inc. Nanopore-based nucleic acid analysis with mixed FRET detection
US10544449B2 (en) 2013-06-14 2020-01-28 Pacific Biosciences Of California, Inc. Bis-biotinylation tags
WO2014201265A1 (en) 2013-06-14 2014-12-18 Pacific Biosciences Of California, Inc. Bis-biotinylation tags
EP3013983B1 (en) 2013-06-25 2023-02-15 Prognosys Biosciences, Inc. Spatially encoded biological assays using a microfluidic device
PL3017065T3 (en) 2013-07-01 2019-03-29 Illumina, Inc. Catalyst-free surface functionalization and polymer grafting
ES2628485T3 (en) 2013-07-03 2017-08-03 Illumina, Inc. Sequencing by orthogonal synthesis
US9612245B2 (en) * 2013-07-03 2017-04-04 University Of North Texas Health Science Center At Fort Worth Multiple-pulse pumping for enhanced fluorescence detection and molecular imaging in cells and tissue
US9957291B2 (en) 2013-08-05 2018-05-01 Pacific Biosciences Of California, Inc. Protected fluorescent reagent compounds
WO2015021228A1 (en) 2013-08-08 2015-02-12 Illumina, Inc. Fluidic system for reagent delivery to a flow cell
EP3036359B1 (en) 2013-08-19 2019-10-23 Abbott Molecular Inc. Next-generation sequencing libraries
JP2016539343A (en) 2013-08-30 2016-12-15 イルミナ インコーポレイテッド Manipulating droplets on hydrophilic or mottled hydrophilic surfaces
US9879318B2 (en) 2013-09-06 2018-01-30 Pacific Biosciences Of California, Inc. Methods and compositions for nucleic acid sample preparation
US9416414B2 (en) 2013-10-24 2016-08-16 Pacific Biosciences Of California, Inc. Delaying real-time sequencing
US10540783B2 (en) * 2013-11-01 2020-01-21 Illumina, Inc. Image analysis useful for patterned objects
CA2929596C (en) 2013-11-13 2022-07-05 Nugen Technologies, Inc. Compositions and methods for identification of a duplicate sequencing read
MX2016006455A (en) 2013-11-17 2016-12-09 Quantum-Si Incorporated Active-source-pixel, integrated device for rapid analysis of biological and chemical speciments.
AU2014360530B2 (en) 2013-12-03 2020-09-03 Illumina, Inc. Methods and systems for analyzing image data
CN111118121A (en) 2013-12-05 2020-05-08 生捷科技控股公司 Preparation of patterned arrays
WO2015085274A1 (en) 2013-12-05 2015-06-11 Centrillion Technology Holdings Corporation Methods for sequencing nucleic acids
EP3077430A4 (en) 2013-12-05 2017-08-16 Centrillion Technology Holdings Corporation Modified surfaces
MX360883B (en) 2013-12-10 2018-11-21 Illumina Inc Biosensors for biological or chemical analysis and methods of manufacturing the same.
WO2015095355A2 (en) 2013-12-17 2015-06-25 The Brigham And Women's Hospital, Inc. Detection of an antibody against a pathogen
ES2890078T3 (en) 2013-12-20 2022-01-17 Illumina Inc Conservation of genomic connectivity information in fragmented genomic DNA samples
EP3778890B1 (en) 2013-12-23 2023-12-06 Illumina, Inc. Structured substrates for improving detection of light emissions and methods relating to the same
US9677132B2 (en) 2014-01-16 2017-06-13 Illumina, Inc. Polynucleotide modification on solid support
EP3094742A1 (en) 2014-01-16 2016-11-23 Illumina, Inc. Amplicon preparation and sequencing on solid supports
WO2015126766A1 (en) 2014-02-18 2015-08-27 Illumina, Inc. Methods and compositions for dna profiling
WO2015131107A1 (en) 2014-02-28 2015-09-03 Nugen Technologies, Inc. Reduced representation bisulfite sequencing with diversity adaptors
US10018566B2 (en) 2014-02-28 2018-07-10 Ldip, Llc Partially encapsulated waveguide based sensing chips, systems and methods of use
EP3116651B1 (en) 2014-03-11 2020-04-22 Illumina, Inc. Disposable, integrated microfluidic cartridge and methods of making it
US11060139B2 (en) 2014-03-28 2021-07-13 Centrillion Technology Holdings Corporation Methods for sequencing nucleic acids
SG11201609053YA (en) 2014-04-29 2016-11-29 Illumina Inc Multiplexed single cell gene expression analysis using template switch and tagmentation
ES2876432T3 (en) 2014-05-16 2021-11-12 Illumina Inc Nucleic acid synthesis techniques
CN110038653A (en) 2014-05-27 2019-07-23 伊鲁米那股份有限公司 The system and method for biochemical analysis including underlying instrument and detachable box
US9708655B2 (en) 2014-06-03 2017-07-18 Illumina, Inc. Compositions, systems, and methods for detecting events using tethers anchored to or adjacent to nanopores
US20150353989A1 (en) 2014-06-09 2015-12-10 Illumina Cambridge Limited Sample preparation for nucleic acid amplification
CA3172086A1 (en) 2014-06-13 2015-12-17 Illumina Cambridge Limited Methods and compositions for preparing sequencing libraries
CN106604994B (en) 2014-06-23 2021-12-14 通用医疗公司 Whole genome unbiased identification of DSBs by sequencing evaluation (GUIDE-Seq)
US11155809B2 (en) 2014-06-24 2021-10-26 Bio-Rad Laboratories, Inc. Digital PCR barcoding
US10017759B2 (en) 2014-06-26 2018-07-10 Illumina, Inc. Library preparation of tagged nucleic acid
JP2017521654A (en) 2014-06-27 2017-08-03 アボット・ラボラトリーズAbbott Laboratories Compositions and methods for detecting human pegivirus 2 (HPgV-2)
PL3161154T3 (en) 2014-06-27 2020-10-19 Illumina, Inc. Modified polymerases for improved incorporation of nucleotide analogues
WO2016003814A1 (en) 2014-06-30 2016-01-07 Illumina, Inc. Methods and compositions using one-sided transposition
SG11201700348VA (en) 2014-07-15 2017-02-27 Illumina Inc Biochemically activated electronic device
CA3176503A1 (en) 2014-07-21 2016-01-28 Illumina, Inc Polynucleotide enrichment using crispr-cas systems
CN107075581B (en) 2014-08-06 2022-03-18 纽亘技术公司 Digital measurement by targeted sequencing
MX2017001807A (en) 2014-08-08 2018-02-08 Quantum Si Inc Optical system and assay chip for probing, detecting, and analyzing molecules.
GB201414098D0 (en) 2014-08-08 2014-09-24 Illumina Cambridge Ltd Modified nucleotide linkers
EP3471402B1 (en) 2014-08-08 2023-05-31 Quantum-Si Incorporated Integrated device for temporal binning of received photons
CN112903639B (en) 2014-08-08 2022-09-13 宽腾矽公司 Integrated device with external light source for detection, detection and analysis of molecules
EP3183367B1 (en) 2014-08-19 2019-06-26 Pacific Biosciences Of California, Inc. Compositions and methods for enrichment of nucleic acids
US10435685B2 (en) 2014-08-19 2019-10-08 Pacific Biosciences Of California, Inc. Compositions and methods for enrichment of nucleic acids
EP3183577B1 (en) 2014-08-21 2020-08-19 Illumina Cambridge Limited Reversible surface functionalization
US9606068B2 (en) 2014-08-27 2017-03-28 Pacific Biosciences Of California, Inc. Arrays of integrated analytical devices
WO2016040602A1 (en) 2014-09-11 2016-03-17 Epicentre Technologies Corporation Reduced representation bisulfite sequencing using uracil n-glycosylase (ung) and endonuclease iv
US10633694B2 (en) 2014-09-12 2020-04-28 Illumina, Inc. Compositions, systems, and methods for detecting the presence of polymer subunits using chemiluminescence
EP3194643B1 (en) * 2014-09-17 2020-02-19 Ibis Biosciences, Inc. Sequencing by synthesis using pulse read optics
WO2016044233A1 (en) 2014-09-18 2016-03-24 Illumina, Inc. Methods and systems for analyzing nucleic acid sequencing data
WO2016054096A1 (en) 2014-09-30 2016-04-07 Illumina, Inc. Modified polymerases for improved incorporation of nucleotide analogues
CA2960721C (en) 2014-10-09 2023-09-05 Illumina, Inc. Method and device for separating immiscible liquids to effectively isolate at least one of the liquids
ES2789000T3 (en) 2014-10-10 2020-10-23 Quantapore Inc Nanopore-based polynucleotide analysis with mutually inactivating fluorescent labels
US9897791B2 (en) 2014-10-16 2018-02-20 Illumina, Inc. Optical scanning systems for in situ genetic analysis
BR122021026779B1 (en) 2014-10-17 2023-12-19 Illumina Cambridge Limited CONTIGUITY PRESERVING TRANSPOSON
JP6757316B2 (en) 2014-10-24 2020-09-16 クアンタポール, インコーポレイテッド Efficient optical analysis of polymers using nanostructured arrays
PT3212684T (en) 2014-10-31 2020-02-03 Illumina Cambridge Ltd Novel polymers and dna copolymer coatings
GB201419731D0 (en) 2014-11-05 2014-12-17 Illumina Cambridge Ltd Sequencing from multiple primers to increase data rate and density
DK3215616T3 (en) 2014-11-05 2020-03-02 Illumina Cambridge Ltd Reducing DNA damage during sample preparation and sequencing using siderophore chelators
DK3218511T3 (en) 2014-11-11 2020-07-20 Illumina Cambridge Ltd Methods and arrays for generating and sequencing nucleic acid monoclonal clusters
EP3218513B1 (en) 2014-11-11 2018-10-31 Illumina, Inc. Polynucleotide amplification using crispr-cas systems
US10233490B2 (en) 2014-11-21 2019-03-19 Metabiotech Corporation Methods for assembling and reading nucleic acid sequences from mixed populations
US10616219B2 (en) * 2014-12-11 2020-04-07 FlowJo, LLC Single cell data management and analysis systems and methods
PL3234187T3 (en) 2014-12-15 2021-10-25 Illumina, Inc. Method for single molecular placement on a substrate
US10302972B2 (en) 2015-01-23 2019-05-28 Pacific Biosciences Of California, Inc. Waveguide transmission
WO2016126941A1 (en) 2015-02-04 2016-08-11 Pacific Biosciences Of California, Inc. Multimeric protected fluorescent reagents
KR20210135626A (en) 2015-02-10 2021-11-15 일루미나, 인코포레이티드 The method and the composition for analyzing the cellular constituent
WO2016138427A1 (en) 2015-02-27 2016-09-01 Indx Lifecare, Inc. Waveguide-based detection system with scanning light source
CN113064236B (en) 2015-03-16 2022-11-01 加利福尼亚太平洋生物科学股份有限公司 Integrated device and system for free space optical coupling
US10576471B2 (en) 2015-03-20 2020-03-03 Illumina, Inc. Fluidics cartridge for use in the vertical or substantially vertical position
US9976174B2 (en) 2015-03-24 2018-05-22 Illumina Cambridge Limited Methods, carrier assemblies, and systems for imaging samples for biological or chemical analysis
US10300452B2 (en) 2015-03-24 2019-05-28 Pacific Biosciences Of California, Inc. Methods and compositions for single molecule composition loading
WO2016156845A1 (en) 2015-03-31 2016-10-06 Illumina Cambridge Limited Surface concatamerization of templates
FI3901281T3 (en) 2015-04-10 2023-01-31 Spatially distinguished, multiplex nucleic acid analysis of biological specimens
WO2016168386A1 (en) 2015-04-14 2016-10-20 Illumina, Inc. Structured substrates for improving detection of light emissions and methods relating to the same
US10844428B2 (en) 2015-04-28 2020-11-24 Illumina, Inc. Error suppression in sequenced DNA fragments using redundant reads with unique molecular indices (UMIS)
DK3294911T3 (en) 2015-05-11 2020-11-16 Illumina Inc Platform for discovery and analysis of therapeutic agents
EP3304383B1 (en) 2015-05-26 2021-07-07 Pacific Biosciences of California, Inc. De novo diploid genome assembly and haplotype sequence reconstruction
US10640809B2 (en) 2015-05-29 2020-05-05 Epicentre Technologies Corporation Methods of analyzing nucleic acids
KR102054571B1 (en) 2015-05-29 2019-12-10 일루미나 케임브리지 리미티드 Improved Use of Surface Primers in Clusters
ES2927121T3 (en) 2015-05-29 2022-11-02 Illumina Inc Sample holder and assay system for performing designated reactions
BR112017025723A2 (en) 2015-06-03 2018-08-14 Illumina Inc compositions, systems and methods for sequencing polynucleotides using polymerase-anchored streams adjacent to nanopores
WO2016201111A1 (en) 2015-06-09 2016-12-15 Centrillion Technology Holdings Corporation Methods for sequencing nucleic acids
CN107924027B (en) 2015-06-12 2024-01-23 加利福尼亚太平洋生物科学股份有限公司 Integrated target waveguide device and system for optical coupling
WO2017007757A1 (en) 2015-07-06 2017-01-12 Illumina, Inc. Balanced ac modulation for driving droplet operations electrodes
EP3320111B1 (en) 2015-07-06 2021-05-05 Illumina Cambridge Limited Sample preparation for nucleic acid amplification
WO2017007753A1 (en) 2015-07-07 2017-01-12 Illumina, Inc. Selective surface patterning via nanoimrinting
EP3325648B1 (en) 2015-07-17 2023-03-29 Illumina, Inc. Polymer sheets for sequencing applications
CN108138225B (en) 2015-07-27 2022-10-14 亿明达股份有限公司 Spatial localization of nucleic acid sequence information
CA2984702A1 (en) 2015-07-30 2017-02-02 Illumina, Inc. Orthogonal deblocking of nucleotides
EP3332032B1 (en) 2015-08-06 2023-12-27 Pacific Biosciences Of California, Inc. Integrated circuits for selectively addressing sparsely arranged electronic measurement devices
US11512348B2 (en) 2015-08-14 2022-11-29 Illumina, Inc. Systems and methods using magnetically-responsive sensors for determining a genetic characteristic
WO2017034868A1 (en) 2015-08-24 2017-03-02 Illumina, Inc. In-line pressure accumulator and flow-control system for biological or chemical assays
WO2017040306A1 (en) 2015-08-28 2017-03-09 Illumina, Inc. Nucleic acid sequence analysis from single cells
CN107921432A (en) 2015-09-02 2018-04-17 伊卢米纳剑桥有限公司 Improve the system and method for the droplet manipulation in flow control system
US10450598B2 (en) 2015-09-11 2019-10-22 Illumina, Inc. Systems and methods for obtaining a droplet having a designated concentration of a substance-of-interest
KR20180043369A (en) 2015-09-11 2018-04-27 더 제너럴 하스피탈 코포레이션 Complete call and sequencing of nuclease DSB (FIND-SEQ)
EP3356526B1 (en) 2015-09-30 2021-08-25 The General Hospital Corporation Comprehensive in vitro reporting of cleavage events by sequencing (circle-seq)
US20190217300A1 (en) 2015-10-22 2019-07-18 Illumina, Inc. Filler fluid for fluidic devices
WO2017087696A1 (en) 2015-11-18 2017-05-26 Pacific Biosciences Of California, Inc. Methods and compositions for loading of polymerase complexes
CN108350491A (en) 2015-11-18 2018-07-31 加利福尼亚太平洋生物科学股份有限公司 Nucleic acid is loaded on base material
US10435741B2 (en) 2015-11-19 2019-10-08 Pacific Biosciences Of California, Inc. Compounds and systems for improving signal detection
EP3376997A4 (en) 2015-11-20 2019-04-24 Pacific Biosciences Of California, Inc. Labeled nucleotide analogs, reaction mixtures, and methods and systems for sequencing
US10676788B2 (en) 2015-11-20 2020-06-09 Pacific Biosciences Of California, Inc. Modified nucleotide reagents
US10669299B2 (en) 2015-11-20 2020-06-02 Pacific Biosciences Of California, Inc. Protected dye-labeled reagents
ES2875759T3 (en) 2015-12-01 2021-11-11 Illumina Inc Digital microfluidic system for single cell isolation and analyte characterization
EP3390657B1 (en) 2015-12-17 2020-09-16 Illumina, Inc. Distinguishing methylation levels in complex biological samples
WO2017120531A1 (en) 2016-01-08 2017-07-13 Bio-Rad Laboratories, Inc. Multiple beads per droplet resolution
EP3403073A1 (en) 2016-01-11 2018-11-21 Illumina, Inc. Detection apparatus having a microfluorometer, a fluidic system, and a flow cell latch clamp module
EP4053545A1 (en) 2016-03-24 2022-09-07 Illumina, Inc. Photonic superlattice-based devices and compositions for use in luminescent imaging, and methods of using the same
CN112892622B (en) 2016-03-28 2022-08-30 亿明达股份有限公司 Multiplanar microarrays
CA3016221C (en) 2016-04-07 2021-09-28 Illumina, Inc. Methods and systems for construction of normalized nucleic acid libraries
US11326206B2 (en) 2016-04-07 2022-05-10 Pacific Biosciences Of California, Inc. Methods of quantifying target nucleic acids and identifying sequence variants
CN109690359B (en) 2016-04-22 2021-10-15 伊鲁米那股份有限公司 Photonic structure based devices and compositions for use in luminescent imaging of multiple sites within a pixel and methods of using the same
AU2017258523B2 (en) 2016-04-29 2020-08-13 Pacific Biosciences Of California, Inc. Method of Nucleic Acid sequence determination
US10597643B2 (en) 2016-04-29 2020-03-24 Omniome, Inc. Polymerases engineered to reduce nucleotide-independent DNA binding
CN116397007A (en) 2016-05-11 2023-07-07 伊鲁米那股份有限公司 Polynucleotide enrichment and amplification Using the ARGONAUTE System
AU2017267653B2 (en) 2016-05-18 2021-05-13 Illumina, Inc. Self assembled patterning using patterned Hydrophobic surfaces
US10544457B2 (en) 2016-06-14 2020-01-28 Pacific Biosciences Of California, Inc. Methods and compositions for enriching compositions for polymerase enzyme complexes
US10370701B2 (en) 2016-06-17 2019-08-06 Pacific Biosciences Of California, Inc. Methods and compositions for generating asymmetrically-tagged nucleic acid fragments
WO2018009346A1 (en) 2016-07-05 2018-01-11 Quantapore, Inc. Optically based nanopore sequencing
EP3485032B1 (en) 2016-07-12 2021-02-17 Life Technologies Corporation Compositions and methods for detecting nucleic acid regions
WO2018015318A1 (en) 2016-07-18 2018-01-25 F. Hoffmann-La Roche Ag Method for generating single-stranded circular dna libraries for single molecule sequencing
CN109477142B (en) 2016-07-18 2022-03-22 豪夫迈·罗氏有限公司 Asymmetric templates and asymmetric methods of nucleic acid sequencing
DK3488002T3 (en) 2016-07-22 2021-06-21 Univ Oregon Health & Science SINGLE CELL WEEKEND LIBRARIES AND COMBINATORY INDEXING METHODS FOR PREPARING IT
US10711300B2 (en) 2016-07-22 2020-07-14 Pacific Biosciences Of California, Inc. Methods and compositions for delivery of molecules and complexes to reaction sites
JP6828140B2 (en) 2016-08-15 2021-02-10 オムニオム インコーポレイテッドOmniome, Inc. Methods and systems for sequencing nucleic acids
US11543417B2 (en) 2016-08-29 2023-01-03 Oslo Universitetssykehus Hf ChIP-seq assays
WO2018064116A1 (en) 2016-09-28 2018-04-05 Illumina, Inc. Methods and systems for data compression
IL290157B2 (en) 2016-10-14 2023-09-01 Illumina Inc Cartridge assembly
US10190155B2 (en) 2016-10-14 2019-01-29 Nugen Technologies, Inc. Molecular tag attachment and transfer
CA3038347A1 (en) 2016-10-19 2018-04-26 Illumina, Inc. Methods for chemical ligation of nucleic acids
CA3041645C (en) 2016-10-24 2021-11-02 Geneinfosec, Inc. Concealing information present within nucleic acids
CA3044231A1 (en) 2016-11-16 2018-05-24 Illumina, Inc. Validation methods and systems for sequence variant calls
GB201619458D0 (en) 2016-11-17 2017-01-04 Spatial Transcriptomics Ab Method for spatial tagging and analysing nucleic acids in a biological specimen
WO2018100724A1 (en) * 2016-12-01 2018-06-07 株式会社日立ハイテクノロジーズ Spot array substrate, nucleic acid analysis method, and nucleic acid analysis device
CA3049961A1 (en) 2016-12-09 2018-06-14 The Broad Institute, Inc. Crispr effector system based diagnostics
WO2018118971A1 (en) 2016-12-19 2018-06-28 Bio-Rad Laboratories, Inc. Droplet tagging contiguity preserved tagmented dna
US11512339B2 (en) 2016-12-22 2022-11-29 Illumina, Inc. Arrays including a resin film and a patterned polymer layer
EP3559262A4 (en) 2016-12-22 2020-07-22 Illumina, Inc. Arrays with quality control tracers
ES2936079T3 (en) 2016-12-22 2023-03-14 Illumina Inc Array including sequencing primer and non-sequencing entity
EP3562962B1 (en) 2016-12-30 2022-01-05 Omniome, Inc. Method and system employing distinguishable polymerases for detecting ternary complexes and identifying cognate nucleotides
GB201704754D0 (en) 2017-01-05 2017-05-10 Illumina Inc Kinetic exclusion amplification of nucleic acid libraries
CN116612818A (en) 2017-01-06 2023-08-18 伊鲁米那股份有限公司 Phase shift correction
AU2018208462B2 (en) 2017-01-10 2021-07-29 Pacific Biosciences Of California, Inc. Polymerases engineered to reduce nucleotide-independent DNA binding
CA3045498C (en) 2017-01-17 2021-07-13 Illumina, Inc. Oncogenic splice variant determination
CN110313034B (en) 2017-01-18 2023-06-06 伊鲁米那股份有限公司 Method, machine-readable medium and computer system for sequencing nucleic acid molecules
CN110177884B (en) 2017-01-20 2021-08-06 欧姆尼欧美公司 Genotyping by polymerase binding
US10975427B2 (en) 2017-01-20 2021-04-13 Omniome, Inc. Process for cognate nucleotide detection in a nucleic acid sequencing workflow
US9932631B1 (en) 2017-09-11 2018-04-03 Omniome, Inc. Genotyping by polymerase binding
AU2017394644B2 (en) 2017-01-20 2020-02-06 Pacific Biosciences Of California, Inc. Allele-specific capture of nucleic acids
DK3354746T3 (en) 2017-01-30 2019-09-02 Gmi Gregor Mendel Inst Fuer Molekulare Pflanzenbiologie Gmbh NEW SPIKE-IN NUCLEOTIDES FOR NORMALIZING SEQUENCE DATA
GB201701688D0 (en) 2017-02-01 2017-03-15 Illumia Inc System and method with fiducials in non-recliner layouts
GB201701689D0 (en) 2017-02-01 2017-03-15 Illumia Inc System and method with fiducials of non-closed shapes
GB201701686D0 (en) 2017-02-01 2017-03-15 Illunina Inc System & method with fiducials having offset layouts
WO2018152162A1 (en) 2017-02-15 2018-08-23 Omniome, Inc. Distinguishing sequences by detecting polymerase dissociation
MX2018015529A (en) 2017-02-21 2019-05-16 Illumina Inc Tagmentation using immobilized transposomes with linkers.
CA3056411A1 (en) 2017-03-15 2018-09-20 The Broad Institute, Inc. Crispr effector system based diagnostics for virus detection
US11021740B2 (en) 2017-03-15 2021-06-01 The Broad Institute, Inc. Devices for CRISPR effector system based diagnostics
US11104937B2 (en) 2017-03-15 2021-08-31 The Broad Institute, Inc. CRISPR effector system based diagnostics
US11174515B2 (en) 2017-03-15 2021-11-16 The Broad Institute, Inc. CRISPR effector system based diagnostics
CN110612351B (en) 2017-03-20 2023-08-11 Illumina公司 Methods and compositions for preparing nucleic acid libraries
WO2018187013A1 (en) 2017-04-04 2018-10-11 Omniome, Inc. Fluidic apparatus and methods useful for chemical and biological reactions
DK3615690T3 (en) 2017-04-23 2021-11-15 Illumina Cambridge Ltd COMPOSITIONS AND METHODS FOR IMPROVING SAMPLE IDENTIFICATION IN INDEXED NUCLEIC ACID LIBRARIES
CA3059839C (en) 2017-04-23 2023-01-03 Illumina Cambridge Limited Compositions and methods for improving sample identification in indexed nucleic acid libraries
ES2937929T3 (en) 2017-04-23 2023-04-03 Illumina Inc Compositions and methods to improve sample identification in indexed nucleic acid libraries
US10161003B2 (en) 2017-04-25 2018-12-25 Omniome, Inc. Methods and apparatus that increase sequencing-by-binding efficiency
CA3220983A1 (en) 2017-05-01 2018-11-08 Illumina, Inc. Optimal index sequences for multiplex massively parallel sequencing
CN110832087A (en) 2017-05-08 2020-02-21 伊鲁米那股份有限公司 Universal short adaptors for indexing of polynucleotide samples
EP3631417B1 (en) 2017-05-25 2024-02-14 Flowjo, LLC Visualization, comparative analysis, and automated difference detection for large multi-parameter data sets
EP3635136B1 (en) 2017-06-07 2021-10-20 Oregon Health & Science University Single cell whole genome libraries for methylation sequencing
CN110770356A (en) 2017-06-20 2020-02-07 生物辐射实验室股份有限公司 MDA using bead oligonucleotides
US11702654B2 (en) 2017-06-20 2023-07-18 Illumina, Inc. Methods and compositions for addressing inefficiencies in amplification reactions
CN111356774B (en) 2017-06-26 2024-01-26 维也纳自然资源与生命科学大学 Novel biomarkers for detecting senescent cells
AU2018302034B2 (en) 2017-07-18 2021-05-20 Pacific Biosciences Of California, Inc. Method of chemically modifying plastic surfaces
WO2019027767A1 (en) 2017-07-31 2019-02-07 Illumina Inc. Sequencing system with multiplexed biological sample aggregation
SG11201911869XA (en) 2017-08-01 2020-01-30 Illumina Inc Spatial indexing of genetic material and library preparation using hydrogel beads and flow cells
EP3545106B1 (en) 2017-08-01 2022-01-19 Helitec Limited Methods of enriching and determining target nucleotide sequences
NZ759924A (en) 2017-08-01 2023-07-28 Illumina Inc Hydrogel beads for nucleotide sequencing
CA3072136A1 (en) 2017-08-15 2019-02-21 Omniome, Inc. Scanning apparatus and methods useful for detection of chemical and biological analytes
CN111315895A (en) 2017-09-14 2020-06-19 豪夫迈·罗氏有限公司 Novel method for generating circular single-stranded DNA library
EP3682027A1 (en) 2017-09-15 2020-07-22 H. Hoffnabb-La Roche Ag Hybridization-extension-ligation strategy for generating circular single-stranded dna libraries
US11447818B2 (en) 2017-09-15 2022-09-20 Illumina, Inc. Universal short adapters with variable length non-random unique molecular identifiers
WO2019060640A1 (en) 2017-09-20 2019-03-28 Guardant Health, Inc. Methods and systems for differentiating somatic and germline variants
WO2019068797A1 (en) 2017-10-06 2019-04-11 F. Hoffmann-La Roche Ag Circularization methods for single molecule sequencing sample preparation
NZ759846A (en) 2017-10-16 2022-05-27 Illumina Inc Deep learning-based splice site classification
IL299565B1 (en) 2017-10-16 2024-03-01 Illumina Inc Recurrent neural network-based variant pathogenicity classifier
US11099202B2 (en) 2017-10-20 2021-08-24 Tecan Genomics, Inc. Reagent delivery system
US11162138B2 (en) 2017-10-30 2021-11-02 Pacific Biosciences Of California, Inc. Multi-amplitude modular labeled compounds
EP4180534A1 (en) 2017-11-02 2023-05-17 Bio-Rad Laboratories, Inc. Transposase-based genomic analysis
WO2019086531A1 (en) 2017-11-03 2019-05-09 F. Hoffmann-La Roche Ag Linear consensus sequencing
US11739367B2 (en) 2017-11-08 2023-08-29 Twinstrand Biosciences, Inc. Reagents and adapters for nucleic acid sequencing and methods for making such reagents and adapters
JP6857235B2 (en) * 2017-11-21 2021-04-14 日本碍子株式会社 Method for manufacturing optical waveguide structure, phosphor element and optical waveguide structure
US10655168B2 (en) 2017-12-22 2020-05-19 Pacific Biosciences Of California, Inc. Modified biotin-binding proteins for immobilization
US11561196B2 (en) 2018-01-08 2023-01-24 Illumina, Inc. Systems and devices for high-throughput sequencing with semiconductor-based detection
ES2964024T3 (en) 2018-01-08 2024-04-03 Illumina Inc High-throughput sequencing with semiconductor-based detection
WO2019140402A1 (en) 2018-01-15 2019-07-18 Illumina, Inc. Deep learning-based variant classifier
FI3746568T3 (en) 2018-01-29 2023-12-12 Broad Inst Inc Crispr effector system based diagnostics
CN111699253A (en) 2018-01-31 2020-09-22 生物辐射实验室股份有限公司 Methods and compositions for deconvolving a partitioned barcode
WO2019149958A1 (en) 2018-02-05 2019-08-08 F. Hoffmann-La Roche Ag Generation of single-stranded circular dna templates for single molecule
CA3089267A1 (en) 2018-02-06 2019-08-15 Omniome, Inc. Compositions and techniques for nucleic acid primer extension
CA3186025A1 (en) 2018-02-13 2019-08-22 Illumina, Inc. Dna sequencing using hydrogel beads
WO2019161253A1 (en) 2018-02-16 2019-08-22 Ultima Genomics, Inc. Methods for sequencing with single frequency detection
JP2021514651A (en) 2018-03-02 2021-06-17 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft Preparation of single-stranded circular DNA template for single molecule sequencing
US11360027B2 (en) 2018-03-29 2022-06-14 Illumina, Inc. Illumination for fluorescence imaging using objective lens
WO2019195225A1 (en) 2018-04-02 2019-10-10 Illumina, Inc. Compositions and methods for making controls for sequence-based genetic testing
WO2019200338A1 (en) 2018-04-12 2019-10-17 Illumina, Inc. Variant classifier based on deep neural networks
US11512002B2 (en) 2018-04-18 2022-11-29 University Of Virginia Patent Foundation Silica materials and methods of making thereof
WO2019203986A1 (en) 2018-04-19 2019-10-24 Omniome, Inc. Improving accuracy of base calls in nucleic acid sequencing methods
SG11201911961RA (en) 2018-04-20 2020-01-30 Illumina Inc Methods of encapsulating single cells, the encapsulated cells and uses thereof
WO2019209426A1 (en) 2018-04-26 2019-10-31 Omniome, Inc. Methods and compositions for stabilizing nucleic acid-nucleotide-polymerase complexes
MX2019015174A (en) 2018-05-15 2023-01-02 Illumina Inc Compositions and methods for chemical cleavage and deprotection of surface-bound oligonucleotides.
CN112534063A (en) 2018-05-22 2021-03-19 安序源有限公司 Methods, systems, and compositions for nucleic acid sequencing
EP3802880A1 (en) 2018-05-25 2021-04-14 Illumina, Inc. Circulating rna signatures specific to preeclampsia
US11180794B2 (en) 2018-05-31 2021-11-23 Omniome, Inc. Methods and compositions for capping nucleic acids
US11339428B2 (en) 2018-05-31 2022-05-24 Pacific Biosciences Of California, Inc. Increased signal to noise in nucleic acid sequencing
MX2019015262A (en) 2018-06-04 2023-01-25 Illumina Inc High-throughput single-cell transcriptome libraries and methods of making and of using.
EP3802878A1 (en) 2018-06-04 2021-04-14 Guardant Health, Inc. Methods and systems for determining the cellular origin of cell-free nucleic acids
EP3814531A4 (en) 2018-06-29 2022-04-06 Pacific Biosciences Of California, Inc. Methods and compositions for delivery of molecules and complexes to reaction sites
US20200251183A1 (en) 2018-07-11 2020-08-06 Illumina, Inc. Deep Learning-Based Framework for Identifying Sequence Patterns that Cause Sequence-Specific Errors (SSEs)
JP2021530219A (en) 2018-07-12 2021-11-11 ツインストランド・バイオサイエンシズ・インコーポレイテッドTwinstrand Biosciences, Inc. Methods and Reagents for Characterizing Genome Editing, Cloning, and Related Applications
WO2020023420A2 (en) 2018-07-23 2020-01-30 Guardant Health, Inc. Methods and systems for adjusting tumor mutational burden by tumor fraction and coverage
CA3107165A1 (en) 2018-07-24 2020-01-30 Omniome, Inc. Serial formation of ternary complex species
CN113286884A (en) 2018-08-07 2021-08-20 博德研究所 Novel CAS12B enzymes and systems
JP7431802B2 (en) 2018-08-15 2024-02-15 イルミナ インコーポレイテッド Compositions and methods for improving library enrichment
US11479816B2 (en) 2018-08-20 2022-10-25 Bio-Rad Laboratories, Inc. Nucleotide sequence generation by barcode bead-colocalization in partitions
CA3109646A1 (en) 2018-08-30 2020-03-05 Guardant Health, Inc. Methods and systems for detecting contamination between samples
CA3109539A1 (en) 2018-08-31 2020-03-05 Guardant Health, Inc. Microsatellite instability detection in cell-free dna
US20200075124A1 (en) 2018-09-04 2020-03-05 Guardant Health, Inc. Methods and systems for detecting allelic imbalance in cell-free nucleic acid samples
EP3853358A1 (en) 2018-09-17 2021-07-28 Omniome, Inc. Engineered polymerases for improved sequencing
SG11202101798QA (en) 2018-09-20 2021-04-29 Tamirna Gmbh Micro-rna signatures for the prediction of liver dysfunction
WO2020072816A1 (en) 2018-10-03 2020-04-09 The Broad Institute, Inc. Crispr effector system based diagnostics for hemorrhagic fever detection
SG10202108013QA (en) 2018-10-15 2021-09-29 Illumina Inc Deep learning-based techniques for pre-training deep convolutional neural networks
BR112021005976A2 (en) 2018-10-26 2021-06-29 Illumina, Inc. modulation of polymer microspheres for DNA processing
CA3116176A1 (en) 2018-10-31 2020-05-07 Guardant Health, Inc. Methods, compositions and systems for calibrating epigenetic partitioning assays
CA3103719A1 (en) 2018-10-31 2020-05-07 Illumina, Inc. Polymerases, compositions, and methods of use
NL2022043B1 (en) 2018-11-21 2020-06-03 Akershus Univ Hf Tagmentation-Associated Multiplex PCR Enrichment Sequencing
CN113166192A (en) 2018-11-30 2021-07-23 吉内恩福赛克公司 Method for generating random oligonucleotides and determining the sequence thereof
BR112021006183A2 (en) 2018-11-30 2021-06-29 Illumina, Inc. analysis of multiple analytes using a single assay
CN113166805A (en) 2018-12-04 2021-07-23 欧姆尼欧美公司 Mixed phase fluids for nucleic acid sequencing and other analytical assays
US11001816B2 (en) 2018-12-05 2021-05-11 Illumina, Inc. Polymerases, compositions, and methods of use
CA3103736A1 (en) 2018-12-05 2020-06-11 Illumina Cambridge Limited Methods and compositions for cluster generation by bridge amplification
CN113454217A (en) 2018-12-07 2021-09-28 奥科坦特公司 System for screening protein-protein interaction
GB201820300D0 (en) 2018-12-13 2019-01-30 10X Genomics Inc Method for spatial tagging and analysing genomic DNA in a biological specimen
GB201820341D0 (en) 2018-12-13 2019-01-30 10X Genomics Inc Method for transposase-mediated spatial tagging and analysing genomic DNA in a biological specimen
CN112639126A (en) 2018-12-14 2021-04-09 伊卢米纳剑桥有限公司 Reducing phasing with unlabeled nucleotides during sequencing
CA3103744A1 (en) 2018-12-17 2020-06-25 Pietro GATTI-LAFRANCONI Compositions for use in polynucleotide sequencing
JP2022512265A (en) 2018-12-17 2022-02-03 イルミナ ケンブリッジ リミテッド Primer oligonucleotides for sequencing
AU2019411272A1 (en) 2018-12-18 2021-01-07 Illumina Cambridge Limited Methods and compositions for paired end sequencing using a single surface primer
DK3899037T3 (en) 2018-12-19 2023-11-06 Illumina Inc METHODS OF IMPROVING POLYNUCLEOTIDE CLUSTER CLONALITY PRIORITY
CN113227348A (en) 2018-12-20 2021-08-06 欧姆尼欧美公司 Temperature control for analysis of nucleic acids and other analytes
US20200232010A1 (en) 2018-12-20 2020-07-23 Guardant Health, Inc. Methods, compositions, and systems for improving recovery of nucleic acid molecules
US11293061B2 (en) 2018-12-26 2022-04-05 Illumina Cambridge Limited Sequencing methods using nucleotides with 3′ AOM blocking group
EP3674702A1 (en) 2018-12-27 2020-07-01 Imec VZW Method for sequencing a polynucleotide using a biofet
US11926867B2 (en) 2019-01-06 2024-03-12 10X Genomics, Inc. Generating capture probes for spatial analysis
US11649485B2 (en) 2019-01-06 2023-05-16 10X Genomics, Inc. Generating capture probes for spatial analysis
SG11202102918WA (en) 2019-01-11 2021-04-29 Illumina Cambridge Ltd Complex surface-bound transposome complexes
CN113661249A (en) 2019-01-31 2021-11-16 夸登特健康公司 Compositions and methods for isolating cell-free DNA
EP3924513B1 (en) 2019-02-14 2023-04-12 Pacific Biosciences of California, Inc. Mitigating adverse impacts of detection systems on nucleic acids and other biological analytes
CN113613787B (en) 2019-02-20 2023-06-13 加利福尼亚太平洋生物科学股份有限公司 Scanning device and method for detecting chemical and biological analytes
WO2020176659A1 (en) 2019-02-27 2020-09-03 Guardant Health, Inc. Methods and systems for determining the cellular origin of cell-free dna
CA3131682A1 (en) 2019-02-28 2020-09-03 Pacific Biosciences Of California, Inc. Improved alignment using homopolymer-collapsed sequencing reads
KR20210134598A (en) 2019-03-01 2021-11-10 일루미나, 인코포레이티드 High-throughput single-nuclear and single-cell libraries and methods of making and using same
NL2023312B1 (en) 2019-03-21 2020-09-28 Illumina Inc Artificial intelligence-based base calling
NL2023314B1 (en) 2019-03-21 2020-09-28 Illumina Inc Artificial intelligence-based quality scoring
NL2023311B9 (en) 2019-03-21 2021-03-12 Illumina Inc Artificial intelligence-based generation of sequencing metadata
NL2023316B1 (en) 2019-03-21 2020-09-28 Illumina Inc Artificial intelligence-based sequencing
WO2020191390A2 (en) 2019-03-21 2020-09-24 Illumina, Inc. Artificial intelligence-based quality scoring
NL2023310B1 (en) 2019-03-21 2020-09-28 Illumina Inc Training data generation for artificial intelligence-based sequencing
US11347965B2 (en) 2019-03-21 2022-05-31 Illumina, Inc. Training data generation for artificial intelligence-based sequencing
US11210554B2 (en) 2019-03-21 2021-12-28 Illumina, Inc. Artificial intelligence-based generation of sequencing metadata
CN109827918B (en) * 2019-04-03 2021-08-13 广西壮族自治区冶金产品质量检验站 Method for measuring silicon content in tin-doped indium oxide powder
US11593649B2 (en) 2019-05-16 2023-02-28 Illumina, Inc. Base calling using convolutions
WO2020243164A1 (en) 2019-05-28 2020-12-03 Octant, Inc. Transcriptional relay system
US11939636B2 (en) 2019-05-31 2024-03-26 Guardant Health, Inc. Methods and systems for improving patient monitoring after surgery
US11644406B2 (en) 2019-06-11 2023-05-09 Pacific Biosciences Of California, Inc. Calibrated focus sensing
CA3104755A1 (en) 2019-06-28 2020-12-30 Illumina Cambridge Limited Flowcells with linear waveguides
CN113226519A (en) 2019-07-12 2021-08-06 Illumina剑桥有限公司 Preparation of nucleic acid libraries using electrophoresis
US20220154173A1 (en) 2019-07-12 2022-05-19 Iiiumina Cambridge Limited Compositions and Methods for Preparing Nucleic Acid Sequencing Libraries Using CRISPR/CAS9 Immobilized on a Solid Support
US11377655B2 (en) 2019-07-16 2022-07-05 Pacific Biosciences Of California, Inc. Synthetic nucleic acids having non-natural structures
US10656368B1 (en) 2019-07-24 2020-05-19 Omniome, Inc. Method and system for biological imaging using a wide field objective lens
TW202124406A (en) 2019-09-10 2021-07-01 美商歐姆尼歐美公司 Reversible modification of nucleotides
WO2021050962A1 (en) 2019-09-11 2021-03-18 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Cancer detection and classification
AU2020361681A1 (en) 2019-10-10 2022-05-05 1859, Inc. Methods and systems for microfluidic screening
EP4045683A1 (en) 2019-10-18 2022-08-24 Omniome, Inc. Methods and compositions for capping nucleic acids
US20210139867A1 (en) 2019-11-08 2021-05-13 Omniome, Inc. Engineered polymerases for improved sequencing by binding
AU2020387423A1 (en) 2019-11-22 2021-09-30 Illumina, Inc. Circulating RNA signatures specific to preeclampsia
WO2021108708A1 (en) 2019-11-26 2021-06-03 Guardant Health, Inc. Methods, compositions and systems for improving the binding of methylated polynucleotides
DE202019106694U1 (en) 2019-12-02 2020-03-19 Omniome, Inc. System for sequencing nucleic acids in fluid foam
DE202019106695U1 (en) 2019-12-02 2020-03-19 Omniome, Inc. System for sequencing nucleic acids in fluid foam
CA3131632A1 (en) 2019-12-04 2021-06-10 Tong Liu Preparation of dna sequencing libraries for detection of dna pathogens in plasma
CA3164781A1 (en) 2019-12-18 2021-06-24 F. Hoffmann-La Roche Ag Methods of sequencing by synthesis using a consecutive labeling scheme
US20220356461A1 (en) 2019-12-19 2022-11-10 Illumina, Inc. High-throughput single-cell libraries and methods of making and of using
US11821035B1 (en) 2020-01-29 2023-11-21 10X Genomics, Inc. Compositions and methods of making gene expression libraries
WO2021152586A1 (en) 2020-01-30 2021-08-05 Yeda Research And Development Co. Ltd. Methods of analyzing microbiome, immunoglobulin profile and physiological state
CN115243792A (en) 2020-02-04 2022-10-25 加利福尼亚太平洋生物科学股份有限公司 Flow cell and methods of making and using the same
US20210265018A1 (en) 2020-02-20 2021-08-26 Illumina, Inc. Knowledge Distillation and Gradient Pruning-Based Compression of Artificial Intelligence-Based Base Caller
US20210265015A1 (en) 2020-02-20 2021-08-26 Illumina, Inc. Hardware Execution and Acceleration of Artificial Intelligence-Based Base Caller
IL295560A (en) 2020-02-20 2022-10-01 Illumina Inc Artificial intelligence-based many-to-many base calling
EP4114966A1 (en) 2020-03-03 2023-01-11 Pacific Biosciences Of California, Inc. Methods and compositions for sequencing double stranded nucleic acids
CA3176615A1 (en) 2020-03-30 2021-10-07 Illumina, Inc. Methods and compositions for preparing nucleic acid libraries
WO2021214766A1 (en) 2020-04-21 2021-10-28 Yeda Research And Development Co. Ltd. Methods of diagnosing viral infections and vaccines thereto
EP4143338A1 (en) 2020-04-30 2023-03-08 Guardant Health, Inc. Methods for sequence determination using partitioned nucleic acids
US20230203592A1 (en) 2020-05-05 2023-06-29 Akershus Universitetssykehus Hf Compositions and methods for characterizing bowel cancer
EP4146822A1 (en) 2020-05-05 2023-03-15 Pacific Biosciences of California, Inc. Compositions and methods for modifying polymerase-nucleic acid complexes
US11188778B1 (en) 2020-05-05 2021-11-30 Illumina, Inc. Equalization-based image processing and spatial crosstalk attenuator
AU2021271637A1 (en) 2020-05-12 2022-12-08 Illumina Singapore Pte. Ltd. Generating nucleic acids with modified bases using recombinant terminal deoxynucleotidyl transferase
WO2021231921A1 (en) 2020-05-14 2021-11-18 Guardant Health, Inc. Homologous recombination repair deficiency detection
WO2021252617A1 (en) 2020-06-09 2021-12-16 Illumina, Inc. Methods for increasing yield of sequencing libraries
EP4165549A1 (en) 2020-06-11 2023-04-19 Nautilus Biotechnology, Inc. Methods and systems for computational decoding of biological, chemical, and physical entities
JP2023531009A (en) 2020-06-22 2023-07-20 イルミナ ケンブリッジ リミテッド Nucleosides and nucleotides with 3' acetal blocking groups
CN111665617A (en) * 2020-06-24 2020-09-15 武汉中纪生物科技有限公司 Focusing method and system
IL299052A (en) 2020-07-02 2023-02-01 Illumina Inc A method to calibrate nucleic acid library seeding efficiency in flowcells
IL299042A (en) 2020-07-08 2023-02-01 Illumina Inc Beads as transposome carriers
WO2023282916A1 (en) 2021-07-09 2023-01-12 Guardant Health, Inc. Methods of detecting genomic rearrangements using cell free nucleic acids
WO2022026761A1 (en) 2020-07-30 2022-02-03 Guardant Health, Inc. Methods for isolating cell-free dna
AU2021320307A1 (en) 2020-08-06 2023-02-16 Illumina Cambridge Limited Preparation of RNA and DNA sequencing libraries using bead-linked transposomes
IL299783A (en) 2020-08-18 2023-03-01 Illumina Inc Sequence-specific targeted transposition and selection and sorting of nucleic acids
JP2023540221A (en) 2020-08-25 2023-09-22 ガーダント ヘルス, インコーポレイテッド Methods and systems for predicting variant origin
US20220067489A1 (en) 2020-08-28 2022-03-03 Illumina, Inc. Detecting and Filtering Clusters Based on Artificial Intelligence-Predicted Base Calls
US11200446B1 (en) 2020-08-31 2021-12-14 Element Biosciences, Inc. Single-pass primary analysis
WO2022053610A1 (en) 2020-09-11 2022-03-17 Illumina Cambridge Limited Methods of enriching a target sequence from a sequencing library using hairpin adaptors
CA3193183A1 (en) 2020-09-30 2022-04-07 Andrew Kennedy Compositions and methods for analyzing dna using partitioning and a methylation-dependent nuclease
JP2023547366A (en) 2020-10-21 2023-11-10 イルミナ インコーポレイテッド Sequencing templates containing multiple inserts and compositions and methods for improving sequencing throughput
US20220162680A1 (en) * 2020-11-20 2022-05-26 Illumina, Inc. Compositions and methods for sequencing using fluorophores and quenchers or donors
EP4267757A1 (en) 2020-12-23 2023-11-01 Guardant Health, Inc. Methods and systems for analyzing methylated polynucleotides
CN112812954A (en) * 2020-12-29 2021-05-18 中国科学院长春光学精密机械与物理研究所 Gene sequencing chip
US20220298568A1 (en) 2021-01-13 2022-09-22 Pacific Biosciences Of California, Inc. Surface structuring with colloidal assembly
CA3203535A1 (en) 2021-01-21 2022-07-28 Gregory KAPP Systems and methods for biomolecule preparation
WO2022165188A1 (en) 2021-01-29 2022-08-04 Illumina, Inc. Methods, compositions and kits to improve seeding efficiency of flow cells with polynucleotides
EP4288562A1 (en) 2021-02-04 2023-12-13 Illumina, Inc. Long indexed-linked read generation on transposome bound beads
US20240043915A1 (en) 2021-02-13 2024-02-08 The General Hospital Corporation Methods and compositions for in situ macromolecule detection and uses thereof
AU2022231055A1 (en) 2021-03-05 2023-09-14 Guardant Health, Inc. Methods and related aspects for analyzing molecular response
EP4305200A1 (en) 2021-03-09 2024-01-17 Guardant Health, Inc. Detecting the presence of a tumor based on off-target polynucleotide sequencing data
KR20230169143A (en) 2021-03-11 2023-12-15 노틸러스 서브시디어리, 인크. Systems and methods for retaining biomolecules
WO2022197752A1 (en) 2021-03-16 2022-09-22 Illumina, Inc. Tile location and/or cycle based weight set selection for base calling
AU2022245985A1 (en) 2021-03-22 2023-09-21 Illumina Cambridge Limited Methods for improving nucleic acid cluster clonality
IL307195A (en) 2021-03-29 2023-11-01 Illumina Inc Improved methods of library preparation
BR112023019894A2 (en) 2021-03-29 2023-11-14 Illumina Inc COMPOSITIONS AND METHODS FOR ASSESSING DNA DAMAGE IN A LIBRARY AND NORMALIZING AMPLICON SIZE DISTORTION
KR20230161955A (en) 2021-03-30 2023-11-28 일루미나, 인코포레이티드 Improved methods for isothermal complementary DNA and library preparation
WO2022212180A1 (en) 2021-03-31 2022-10-06 Illumina, Inc. Artificial intelligence-based base caller with contextual awareness
AU2022249289A1 (en) 2021-03-31 2023-08-17 Illumina Cambridge Limited Methods of preparing directional tagmentation sequencing libraries using transposon-based technology with unique molecular identifiers for error correction
WO2022208171A1 (en) 2021-03-31 2022-10-06 UCL Business Ltd. Methods for analyte detection
JP2024512651A (en) 2021-04-02 2024-03-19 イルミナ インコーポレイテッド Nucleotides for Sequencing - Machine Learning Model for Detecting Bubbles in Sample Slides
US20220336054A1 (en) 2021-04-15 2022-10-20 Illumina, Inc. Deep Convolutional Neural Networks to Predict Variant Pathogenicity using Three-Dimensional (3D) Protein Structures
US20220356515A1 (en) 2021-05-10 2022-11-10 Pacific Biosciences Of California, Inc. Dna amplification buffer replenishment during rolling circle amplification
US20220356519A1 (en) 2021-05-10 2022-11-10 Pacific Biosciences Of California, Inc. Single-molecule seeding and amplification on a surface
EP4341434A1 (en) 2021-05-20 2024-03-27 Illumina, Inc. Compositions and methods for sequencing by synthesis
WO2022265994A1 (en) 2021-06-15 2022-12-22 Illumina, Inc. Hydrogel-free surface functionalization for sequencing
WO2022272260A1 (en) 2021-06-23 2022-12-29 Illumina, Inc. Compositions, methods, kits, cartridges, and systems for sequencing reagents
WO2023278608A1 (en) 2021-06-29 2023-01-05 Illumina, Inc. Self-learned base caller, trained using oligo sequences
WO2023278184A1 (en) 2021-06-29 2023-01-05 Illumina, Inc. Methods and systems to correct crosstalk in illumination emitted from reaction sites
AU2022305321A1 (en) 2021-06-29 2024-01-18 Illumina, Inc. Signal-to-noise-ratio metric for determining nucleotide-base calls and base-call quality
US20220415443A1 (en) 2021-06-29 2022-12-29 Illumina, Inc. Machine-learning model for generating confidence classifications for genomic coordinates
US20230027409A1 (en) 2021-07-13 2023-01-26 Illumina, Inc. Methods and systems for real time extraction of crosstalk in illumination emitted from reaction sites
WO2023003757A1 (en) 2021-07-19 2023-01-26 Illumina Software, Inc. Intensity extraction with interpolation and adaptation for base calling
US11455487B1 (en) 2021-10-26 2022-09-27 Illumina Software, Inc. Intensity extraction and crosstalk attenuation using interpolation and adaptation for base calling
US20230021577A1 (en) 2021-07-23 2023-01-26 Illumina Software, Inc. Machine-learning model for recalibrating nucleotide-base calls
US20230116852A1 (en) 2021-07-23 2023-04-13 Illumina, Inc. Methods for preparing substrate surface for dna sequencing
WO2023009758A1 (en) 2021-07-28 2023-02-02 Illumina, Inc. Quality score calibration of basecalling systems
WO2023014741A1 (en) 2021-08-03 2023-02-09 Illumina Software, Inc. Base calling using multiple base caller models
US20230047225A1 (en) 2021-08-14 2023-02-16 Illumina, Inc. Polymerases, compositions, and methods of use
CA3223390A1 (en) 2021-08-17 2023-02-23 Colin Brown Methods and compositions for identifying methylated cytosines
US20230070896A1 (en) 2021-09-09 2023-03-09 Nautilus Biotechnology, Inc. Characterization and localization of protein modifications
US20230093253A1 (en) 2021-09-17 2023-03-23 Illumina, Inc. Automatically identifying failure sources in nucleotide sequencing from base-call-error patterns
WO2023049558A1 (en) 2021-09-21 2023-03-30 Illumina, Inc. A graph reference genome and base-calling approach using imputed haplotypes
AU2022352593A1 (en) 2021-09-22 2024-02-15 Nautilus Subsidiary, Inc. Methods and systems for determining polypeptide interactions
WO2023049215A1 (en) 2021-09-22 2023-03-30 Illumina, Inc. Compressed state-based base calling
WO2023056328A2 (en) 2021-09-30 2023-04-06 Illumina, Inc. Solid supports and methods for depleting and/or enriching library fragments prepared from biosamples
US20230096386A1 (en) 2021-09-30 2023-03-30 Illumina Cambridge Limited Polynucleotide sequencing
US20230114905A1 (en) 2021-10-11 2023-04-13 Nautilus Biotechnology, Inc. Highly multiplexable analysis of proteins and proteomes
WO2023069927A1 (en) 2021-10-20 2023-04-27 Illumina, Inc. Methods for capturing library dna for sequencing
US20230149883A1 (en) 2021-11-03 2023-05-18 Nautilus Biotechnology, Inc. Systems and methods for surface structuring
WO2023081485A1 (en) 2021-11-08 2023-05-11 Pacific Biosciences Of California, Inc. Stepwise sequencing of a polynucleotide with a homogenous reaction mixture
US20230343415A1 (en) 2021-12-02 2023-10-26 Illumina Software, Inc. Generating cluster-specific-signal corrections for determining nucleotide-base calls
US20230215515A1 (en) 2021-12-23 2023-07-06 Illumina Software, Inc. Facilitating secure execution of external workflows for genomic sequencing diagnostics
WO2023122363A1 (en) 2021-12-23 2023-06-29 Illumina Software, Inc. Dynamic graphical status summaries for nucelotide sequencing
US20230207050A1 (en) 2021-12-28 2023-06-29 Illumina Software, Inc. Machine learning model for recalibrating nucleotide base calls corresponding to target variants
WO2023129764A1 (en) 2021-12-29 2023-07-06 Illumina Software, Inc. Automatically switching variant analysis model versions for genomic analysis applications
AU2023208743A1 (en) 2022-01-20 2024-01-04 Illumina, Inc. Methods of detecting methylcytosine and hydroxymethylcytosine by sequencing
US20230410944A1 (en) 2022-02-25 2023-12-21 Illumina, Inc. Calibration sequences for nucelotide sequencing
US20230313271A1 (en) 2022-02-25 2023-10-05 Illumina, Inc. Machine-learning models for detecting and adjusting values for nucleotide methylation levels
US20230343414A1 (en) 2022-03-25 2023-10-26 Illumina, Inc. Sequence-to-sequence base calling
WO2023192917A1 (en) 2022-03-29 2023-10-05 Nautilus Subsidiary, Inc. Integrated arrays for single-analyte processes
WO2023196572A1 (en) 2022-04-07 2023-10-12 Illumina Singapore Pte. Ltd. Altered cytidine deaminases and methods of use
WO2023212490A1 (en) 2022-04-25 2023-11-02 Nautilus Subsidiary, Inc. Systems and methods for assessing and improving the quality of multiplex molecular assays
WO2023212601A1 (en) 2022-04-26 2023-11-02 Illumina, Inc. Machine-learning models for selecting oligonucleotide probes for array technologies
WO2023209606A1 (en) 2022-04-29 2023-11-02 Illumina Cambridge Limited Methods and systems for encapsulating lyophilised microspheres
WO2023220602A1 (en) 2022-05-09 2023-11-16 Guardant Health, Inc. Detecting degradation based on strand bias
US20230368866A1 (en) 2022-05-10 2023-11-16 Illumina Software, Inc. Adaptive neural network for nucelotide sequencing
US20230392207A1 (en) 2022-06-03 2023-12-07 Illumina, Inc. Circulating rna biomarkers for preeclampsia
WO2023240093A1 (en) 2022-06-06 2023-12-14 Element Biosciences, Inc. Methods for assembling and reading nucleic acid sequences from mixed populations
WO2023250364A1 (en) 2022-06-21 2023-12-28 Nautilus Subsidiary, Inc. Method for detecting analytes at sites of optically non-resolvable distances
US20230420080A1 (en) 2022-06-24 2023-12-28 Illumina Software, Inc. Split-read alignment by intelligently identifying and scoring candidate split groups
US20230420082A1 (en) 2022-06-27 2023-12-28 Illumina Software, Inc. Generating and implementing a structural variation graph genome
US20230420075A1 (en) 2022-06-27 2023-12-28 Illumina Software, Inc. Accelerators for a genotype imputation model
WO2024006705A1 (en) 2022-06-27 2024-01-04 Illumina Software, Inc. Improved human leukocyte antigen (hla) genotyping
WO2024011145A1 (en) 2022-07-05 2024-01-11 Element Biosciences, Inc. Pcr-free library preparation using double-stranded splint adaptors and methods of use
WO2024015962A1 (en) 2022-07-15 2024-01-18 Pacific Biosciences Of California, Inc. Blocked asymmetric hairpin adaptors
WO2024026356A1 (en) 2022-07-26 2024-02-01 Illumina, Inc. Rapid single-cell multiomics processing using an executable file
WO2024040068A1 (en) 2022-08-15 2024-02-22 Element Biosciences, Inc. Spatially resolved surface capture of nucleic acids
WO2024039516A1 (en) 2022-08-19 2024-02-22 Illumina, Inc. Third dna base pair site-specific dna detection
WO2024059655A1 (en) 2022-09-15 2024-03-21 Nautilus Subsidiary, Inc. Characterizing accessibility of macromolecule structures
WO2024064900A1 (en) 2022-09-22 2024-03-28 Pacific Biosciences Of California, Inc. Systems and methods for tandem repeat mapping

Family Cites Families (115)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4994373A (en) * 1983-01-27 1991-02-19 Enzo Biochem, Inc. Method and structures employing chemically-labelled polynucleotide probes
US5200313A (en) * 1983-08-05 1993-04-06 Miles Inc. Nucleic acid hybridization assay employing detectable anti-hybrid antibodies
CA1338457C (en) 1986-08-22 1996-07-16 Henry A. Erlich Purified thermostable enzyme
US5198543A (en) 1989-03-24 1993-03-30 Consejo Superior Investigaciones Cientificas PHI29 DNA polymerase
US5001050A (en) 1989-03-24 1991-03-19 Consejo Superior Investigaciones Cientificas PHφ29 DNA polymerase
GB8910880D0 (en) 1989-05-11 1989-06-28 Amersham Int Plc Sequencing method
US5302509A (en) * 1989-08-14 1994-04-12 Beckman Instruments, Inc. Method for sequencing polynucleotides
WO1991006678A1 (en) * 1989-10-26 1991-05-16 Sri International Dna sequencing
IL97222A (en) 1990-02-16 1995-08-31 Orion Yhtymae Oy Method and reagent for determining specific nucleotide variations
US5401847A (en) * 1990-03-14 1995-03-28 Regents Of The University Of California DNA complexes with dyes designed for energy transfer as fluorescent markers
DE69132843T2 (en) 1990-12-06 2002-09-12 Affymetrix Inc N D Ges D Staat Identification of nucleic acids in samples
US5455166A (en) * 1991-01-31 1995-10-03 Becton, Dickinson And Company Strand displacement amplification
US6004744A (en) * 1991-03-05 1999-12-21 Molecular Tool, Inc. Method for determining nucleotide identity through extension of immobilized primer
US5255083A (en) * 1991-06-05 1993-10-19 Sony Corporation Of America Digital color correction system and method
US5405747A (en) * 1991-09-25 1995-04-11 The Regents Of The University Of California Office Of Technology Transfer Method for rapid base sequencing in DNA and RNA with two base labeling
US6048690A (en) 1991-11-07 2000-04-11 Nanogen, Inc. Methods for electronic fluorescent perturbation for analysis and electronic perturbation catalysis for synthesis
US5243618A (en) * 1991-11-22 1993-09-07 Hughes Aircraft Company Cavity resonator incorporating waveguide filter
US5221592A (en) * 1992-03-06 1993-06-22 Hoechst Celanese Corporation Diazo ester of a benzolactone ring compound and positive photoresist composition and element utilizing the diazo ester
US5470705A (en) * 1992-04-03 1995-11-28 Applied Biosystems, Inc. Probe composition containing a binding domain and polymer chain and methods of use
GB9208733D0 (en) 1992-04-22 1992-06-10 Medical Res Council Dna sequencing method
US5403708A (en) * 1992-07-06 1995-04-04 Brennan; Thomas M. Methods and compositions for determining the sequence of nucleic acids
US5503980A (en) * 1992-11-06 1996-04-02 Trustees Of Boston University Positional sequencing by hybridization
EP1262564A3 (en) * 1993-01-07 2004-03-31 Sequenom, Inc. Dna sequencing by mass spectrometry
US5465151A (en) * 1993-01-21 1995-11-07 State Of Oregon Acting By And Through The State Board Of Higher Education On Behalf Of The University Of Oregon Sensors employing interference of electromagnetic waves passing through waveguides having functionalized surfaces
US5677196A (en) * 1993-05-18 1997-10-14 University Of Utah Research Foundation Apparatus and methods for multi-analyte homogeneous fluoro-immunoassays
US5874239A (en) 1993-07-30 1999-02-23 Affymax Technologies N.V. Biotinylation of proteins
WO1995006138A1 (en) * 1993-08-25 1995-03-02 The Regents Of The University Of California Microscopic method for detecting micromotions
US5470710A (en) 1993-10-22 1995-11-28 University Of Utah Automated hybridization/imaging device for fluorescent multiplex DNA sequencing
US5654419A (en) * 1994-02-01 1997-08-05 The Regents Of The University Of California Fluorescent labels and their use in separations
US5695934A (en) * 1994-10-13 1997-12-09 Lynx Therapeutics, Inc. Massively parallel sequencing of sorted polynucleotides
US5601982A (en) * 1995-02-07 1997-02-11 Sargent; Jeannine P. Method and apparatus for determining the sequence of polynucleotides
AU5171696A (en) 1995-02-27 1996-09-18 Ely Michael Rabani Device, compounds, algorithms, and methods of molecular characterization and manipulation with molecular parallelism
US5961923A (en) * 1995-04-25 1999-10-05 Irori Matrices with memories and uses thereof
US5677769A (en) * 1995-05-30 1997-10-14 Imra America Optical sensor utilizing rare-earth-doped integrated-optic lasers
EP0745686A1 (en) 1995-06-01 1996-12-04 Roche Diagnostics GmbH The use of DNA polymerase 3'-intrinsic editing activity
US5856174A (en) * 1995-06-29 1999-01-05 Affymetrix, Inc. Integrated nucleic acid diagnostic device
US5661028A (en) 1995-09-29 1997-08-26 Lockheed Martin Energy Systems, Inc. Large scale DNA microsequencing device
US5854033A (en) * 1995-11-21 1998-12-29 Yale University Rolling circle replication reporter systems
US6027890A (en) * 1996-01-23 2000-02-22 Rapigene, Inc. Methods and compositions for enhancing sensitivity in the analysis of biological-based assays
US5846727A (en) 1996-06-06 1998-12-08 Board Of Supervisors Of Louisiana State University And Agricultural & Mechanical College Microsystem for rapid DNA sequencing
US6258533B1 (en) * 1996-11-01 2001-07-10 The University Of Iowa Research Foundation Iterative and regenerative DNA sequencing method
US5858671A (en) * 1996-11-01 1999-01-12 The University Of Iowa Research Foundation Iterative and regenerative DNA sequencing method
US6136543A (en) * 1997-01-31 2000-10-24 Hitachi, Ltd. Method for determining nucleic acids base sequence and apparatus therefor
US5773308A (en) * 1997-02-10 1998-06-30 The United States Of America As Represented By The Secretary Of The Navy Photoactivatable o-nitrobenzyl polyethylene glycol-silane for the production of patterned biomolecular arrays
US6403311B1 (en) * 1997-02-12 2002-06-11 Us Genomics Methods of analyzing polymers using ordered label strategies
DE69825601T2 (en) * 1997-02-12 2005-04-28 Chan, Eugene Y, Brookline METHOD FOR THE ANALYSIS OF POLYMERS
JP2001517948A (en) 1997-04-01 2001-10-09 グラクソ、グループ、リミテッド Nucleic acid sequencing
US5866366A (en) 1997-07-01 1999-02-02 Smithkline Beecham Corporation gidB
KR100664331B1 (en) 1997-07-28 2007-01-02 메디칼 바이오시스템스 리미티드 Nucleic acid sequence analysis
US7348181B2 (en) * 1997-10-06 2008-03-25 Trustees Of Tufts College Self-encoding sensor with microspheres
US6124120A (en) * 1997-10-08 2000-09-26 Yale University Multiple displacement amplification
JP2001519538A (en) 1997-10-10 2001-10-23 プレジデント・アンド・フェローズ・オブ・ハーバード・カレッジ Replica amplification of nucleic acid arrays
US6485944B1 (en) 1997-10-10 2002-11-26 President And Fellows Of Harvard College Replica amplification of nucleic acid arrays
JP3862845B2 (en) * 1998-02-05 2006-12-27 セイコーインスツル株式会社 Near-field optical probe
US6049641A (en) 1998-02-24 2000-04-11 Gemfire Corporation Connection system for optical redundancy
US6326725B1 (en) * 1998-05-26 2001-12-04 Micron Technology, Inc. Focusing electrode for field emission displays and method
GB9815702D0 (en) 1998-07-21 1998-09-16 Cambridge Imaging Ltd Improved imaging system for fluorescence assays
AU770831B2 (en) 1998-07-30 2004-03-04 Solexa Ltd. Arrayed biomolecules and their use in sequencing
US6210896B1 (en) 1998-08-13 2001-04-03 Us Genomics Molecular motors
EP1105535A1 (en) 1998-08-13 2001-06-13 U.S. Genomics Optically characterizing polymers
US6263286B1 (en) * 1998-08-13 2001-07-17 U.S. Genomics, Inc. Methods of analyzing polymers using a spatial network of fluorophores and fluorescence resonance energy transfer
US6280939B1 (en) * 1998-09-01 2001-08-28 Veeco Instruments, Inc. Method and apparatus for DNA sequencing using a local sensitive force detector
EP1114184A2 (en) 1998-09-15 2001-07-11 Yale University Molecular cloning using rolling circle amplification
DE19844931C1 (en) * 1998-09-30 2000-06-15 Stefan Seeger Procedures for DNA or RNA sequencing
US6221592B1 (en) * 1998-10-20 2001-04-24 Wisconsin Alumi Research Foundation Computer-based methods and systems for sequencing of individual nucleic acid molecules
ATE319857T1 (en) 1998-12-14 2006-03-15 Li Cor Inc KIT AND METHOD FOR NUCLEIC ACID SEQUENCING OF INDIVIDUAL MOLECULES BY POLYMERASE SYNTHESIS
GB9828785D0 (en) 1998-12-30 1999-02-17 Amersham Pharm Biotech Ab Sequencing systems
ATE343425T1 (en) * 1999-01-08 2006-11-15 Applera Corp FIBER MATRIX FOR BRINGING CHEMICAL SUBSTANCES TOGETHER, AND METHOD FOR THE PRODUCTION AND USE THEREOF
WO2000041524A2 (en) * 1999-01-11 2000-07-20 President And Fellows Of Harvard College Isothermal amplification of dna
WO2000042233A1 (en) * 1999-01-13 2000-07-20 Cornell Research Foundation, Inc. Monolithic fabrication of fluidic structures
EP1961826A3 (en) 1999-03-10 2008-09-17 ASM Scientific, Inc. A method for direct nucleic acid sequencing
US6515751B1 (en) 1999-03-11 2003-02-04 Cornell Research Foundation Inc. Mechanically resonant nanostructures
WO2000058507A1 (en) 1999-03-30 2000-10-05 Solexa Ltd. Polynucleotide sequencing
US7056661B2 (en) * 1999-05-19 2006-06-06 Cornell Research Foundation, Inc. Method for sequencing nucleic acid molecules
US6818395B1 (en) 1999-06-28 2004-11-16 California Institute Of Technology Methods and apparatus for analyzing polynucleotide sequences
WO2001016375A2 (en) 1999-08-30 2001-03-08 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services High speed parallel molecular nucleic acid sequencing
US6274320B1 (en) * 1999-09-16 2001-08-14 Curagen Corporation Method of sequencing a nucleic acid
US7244559B2 (en) * 1999-09-16 2007-07-17 454 Life Sciences Corporation Method of sequencing a nucleic acid
US6399335B1 (en) 1999-11-16 2002-06-04 Advanced Research And Technology Institute, Inc. γ-phosphoester nucleoside triphosphates
GB0000896D0 (en) * 2000-01-14 2000-03-08 Univ Glasgow Improved analytical chip
US6510263B1 (en) 2000-01-27 2003-01-21 Unaxis Balzers Aktiengesellschaft Waveguide plate and process for its production and microtitre plate
DE10017824B4 (en) * 2000-04-10 2004-03-18 Till I.D. Gmbh Device for parallel photometric fluorescence or luminescence analysis of several separate sample areas on an object
US6291187B1 (en) 2000-05-12 2001-09-18 Molecular Staging, Inc. Poly-primed amplification of nucleic acid sequences
US6917726B2 (en) 2001-09-27 2005-07-12 Cornell Research Foundation, Inc. Zero-mode clad waveguides for performing spectroscopy with confined effective observation volumes
US20030027308A1 (en) * 2000-05-30 2003-02-06 Sugen Incorporated Novel human protein phosphatases identified from genomic sequencing
JP4812223B2 (en) 2000-06-02 2011-11-09 バイエル・テクノロジー・サービシーズ・ゲゼルシャフト・ミット・ベシュレンクテル・ハフツング Kit and method for multi-analyte determination
US6323009B1 (en) 2000-06-28 2001-11-27 Molecular Staging, Inc. Multiply-primed amplification of nucleic acid sequences
CN101525660A (en) * 2000-07-07 2009-09-09 维西根生物技术公司 An instant sequencing methodology
WO2002046357A1 (en) 2000-10-26 2002-06-13 The Trustees Of Princeton University, Princeton University Method and apparatus for dielectric spectroscopy of biological solutions
US20040157306A1 (en) * 2000-11-13 2004-08-12 Sugen Incorporated Mammalian protein phosphatases
GB2374924B (en) * 2001-01-03 2003-12-03 Packard Instrument Co Inc System for alternatively measuring epi-fluorescence or luminescence
DE10102578C2 (en) * 2001-01-20 2003-01-09 Univ Braunschweig Tech Resonant microwave sensor
JP2002237200A (en) * 2001-02-13 2002-08-23 Mitsubishi Electric Corp Semiconductor device and its test method
US7297518B2 (en) * 2001-03-12 2007-11-20 California Institute Of Technology Methods and apparatus for analyzing polynucleotide sequences by asynchronous base extension
US20030143556A1 (en) * 2001-04-03 2003-07-31 Gary Blackburn Nucleic acid reactions using labels with different redox potentials
AU2002303258A1 (en) 2001-04-06 2002-10-28 Incyte Genomics, Inc. Kinases and phosphatases
US20040203097A1 (en) * 2001-05-24 2004-10-14 Henry Yue Kinases and phosphatases
US7118907B2 (en) * 2001-06-06 2006-10-10 Li-Cor, Inc. Single molecule detection systems and methods
EP1417474B1 (en) * 2001-07-25 2021-12-29 The Trustees Of Princeton University Nanochannel arrays and their preparation and use for high throughput macromolecular analysis
ATE458067T1 (en) 2001-08-29 2010-03-15 Ge Healthcare Bio Sciences LABELED NUCLEOSIDE POLYPHOSPHATES
US6617137B2 (en) 2001-10-15 2003-09-09 Molecular Staging Inc. Method of amplifying whole genomes without subjecting the genome to denaturing conditions
US6713672B1 (en) * 2001-12-07 2004-03-30 Laird Technologies, Inc. Compliant shaped EMI shield
JP2003185569A (en) * 2001-12-14 2003-07-03 Mitsubishi Chemicals Corp Analyzer for sample using surface plasmon resonance and sensor chip for surface plasmon resonance analysis
US20030123827A1 (en) * 2001-12-28 2003-07-03 Xtalight, Inc. Systems and methods of manufacturing integrated photonic circuit devices
JP3897703B2 (en) * 2002-01-11 2007-03-28 キヤノン株式会社 Sensor device and inspection method using the same
US6618537B2 (en) 2002-01-14 2003-09-09 Applied Wdm, Inc. Optical waveguide structures and methods of fabrication
DE10217568A1 (en) * 2002-04-19 2003-11-13 Infineon Technologies Ag Waveguides in porous substrates
JP2003344433A (en) * 2002-05-22 2003-12-03 Okutekku:Kk Micro-array, micro-array system and measuring method of test material
US6977153B2 (en) * 2002-12-31 2005-12-20 Qiagen Gmbh Rolling circle amplification of RNA
US20050186317A1 (en) * 2003-06-23 2005-08-25 Richard Dempster Determination of dough development using near infrared radiation
WO2005080605A2 (en) * 2004-02-19 2005-09-01 Helicos Biosciences Corporation Methods and kits for analyzing polynucleotide sequences
US7256886B2 (en) * 2004-07-22 2007-08-14 University Of Maryland At Baltimore County Surface enhanced Raman spectroscopic nano-imaging probe and uses therefor
US7170050B2 (en) 2004-09-17 2007-01-30 Pacific Biosciences Of California, Inc. Apparatus and methods for optical analysis of molecules
US7476503B2 (en) * 2004-09-17 2009-01-13 Pacific Biosciences Of California, Inc. Apparatus and method for performing nucleic acid analysis
US7715001B2 (en) * 2006-02-13 2010-05-11 Pacific Biosciences Of California, Inc. Methods and systems for simultaneous real-time monitoring of optical signals from multiple sources

Also Published As

Publication number Publication date
JP2016154560A (en) 2016-09-01
US20060061754A1 (en) 2006-03-23
CN101914620A (en) 2010-12-15
JP2014057601A (en) 2014-04-03
CN101914620B (en) 2014-02-12
US20060061755A1 (en) 2006-03-23
US7476503B2 (en) 2009-01-13
EP3415641B1 (en) 2023-11-01
GB2423819B (en) 2008-02-06
JP2008513782A (en) 2008-05-01
AU2005296200B2 (en) 2011-07-14
EP3415641A1 (en) 2018-12-19
EP1790202A2 (en) 2007-05-30
WO2006044078A3 (en) 2008-02-21
CA2579150A1 (en) 2006-04-27
US20060062531A1 (en) 2006-03-23
US7302146B2 (en) 2007-11-27
US7315019B2 (en) 2008-01-01
WO2006044078A2 (en) 2006-04-27
AU2005296200A1 (en) 2006-04-27
GB2423819A (en) 2006-09-06
GB0608338D0 (en) 2006-06-07
US20060063264A1 (en) 2006-03-23
JP6294064B2 (en) 2018-03-14
EP1790202A4 (en) 2013-02-20

Similar Documents

Publication Publication Date Title
US9709503B2 (en) Apparatus and method for performing nucleic acid analysis
CA2579150C (en) Apparatus and method for analysis of molecules
US7476504B2 (en) Use of reversible extension terminator in nucleic acid sequencing
US8993230B2 (en) Asynchronous sequencing of biological polymers
US20110217698A1 (en) Method to improve single molecule analyses
AU2011224119B2 (en) Apparatus and method for analysis of molecules

Legal Events

Date Code Title Description
EEER Examination request