US20230338913A1 - Devices and methods for oligonucleic acid library synthesis - Google Patents

Devices and methods for oligonucleic acid library synthesis Download PDF

Info

Publication number
US20230338913A1
US20230338913A1 US18/319,356 US202318319356A US2023338913A1 US 20230338913 A1 US20230338913 A1 US 20230338913A1 US 202318319356 A US202318319356 A US 202318319356A US 2023338913 A1 US2023338913 A1 US 2023338913A1
Authority
US
United States
Prior art keywords
substrate
instances
microchannels
oligonucleic
cases
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US18/319,356
Inventor
William Banyai
Bill James Peck
Andres Fernandez
Siyuan Chen
Pierre Indermuhle
Eugene P. Marsh
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.)
Twist Bioscience Corp
Original Assignee
Twist Bioscience Corp
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Twist Bioscience Corp filed Critical Twist Bioscience Corp
Priority to US18/319,356 priority Critical patent/US20230338913A1/en
Publication of US20230338913A1 publication Critical patent/US20230338913A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J19/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J19/0046Sequential or parallel reactions, e.g. for the synthesis of polypeptides or polynucleotides; Apparatus and devices for combinatorial chemistry or for making molecular arrays
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • C12N15/1034Isolating an individual clone by screening libraries
    • C12N15/1093General methods of preparing gene libraries, not provided for in other subgroups
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00277Apparatus
    • B01J2219/00279Features relating to reactor vessels
    • B01J2219/00306Reactor vessels in a multiple arrangement
    • B01J2219/00313Reactor vessels in a multiple arrangement the reactor vessels being formed by arrays of wells in blocks
    • B01J2219/00315Microtiter plates
    • B01J2219/00317Microwell devices, i.e. having large numbers of wells
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00277Apparatus
    • B01J2219/00497Features relating to the solid phase supports
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00277Apparatus
    • B01J2219/00497Features relating to the solid phase supports
    • B01J2219/00511Walls of reactor vessels
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00277Apparatus
    • B01J2219/00497Features relating to the solid phase supports
    • B01J2219/00513Essentially linear supports
    • B01J2219/0052Essentially linear supports in the shape of elongated tubes
    • B01J2219/00522Essentially linear supports in the shape of elongated tubes in a multiple parallel arrangement
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00585Parallel processes
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00596Solid-phase processes
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00603Making arrays on substantially continuous surfaces
    • B01J2219/00605Making arrays on substantially continuous surfaces the compounds being directly bound or immobilised to solid supports
    • B01J2219/00632Introduction of reactive groups to the surface
    • B01J2219/00635Introduction of reactive groups to the surface by reactive plasma treatment
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00603Making arrays on substantially continuous surfaces
    • B01J2219/00605Making arrays on substantially continuous surfaces the compounds being directly bound or immobilised to solid supports
    • B01J2219/00632Introduction of reactive groups to the surface
    • B01J2219/00637Introduction of reactive groups to the surface by coating it with another layer
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00603Making arrays on substantially continuous surfaces
    • B01J2219/00659Two-dimensional arrays
    • B01J2219/00662Two-dimensional arrays within two-dimensional arrays
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00718Type of compounds synthesised
    • B01J2219/0072Organic compounds
    • B01J2219/00722Nucleotides
    • 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
    • B01L3/50857Containers 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 using arrays or bundles of open capillaries for holding samples

Definitions

  • devices for synthesizing oligonucleic acids comprising: a plate; a main channel, wherein the main channel extends vertically into the plate from an opening on a top side of the plate, and wherein the main channel has a width of 0.5 to 2 mm; and a plurality of microchannels connected to the main channel, wherein each microchannel of the plurality of microchannels extends vertically from an opening on a bottom side of the plate into the main channel, and wherein each microchannel of the plurality of microchannels has a surface area to volume ratio of greater than 0.2 (1/um).
  • the surface area to volume ratio provides for rapid exchange of chemical exposure during de novo synthesis of oligonucleic acids.
  • each microchannel of the plurality of microchannels has a surface area greater than 10,000 um 2 .
  • each microchannel of the plurality of microchannels has a surface area greater than 12,000 um 2 .
  • each microchannel of the plurality of microchannels has a surface area of about 13,000 um 2 .
  • the plurality of microchannels comprises 50 to 500 microchannels.
  • the plurality of microchannels comprises 100 to 150 microchannels. Devices are further provided wherein a ratio of width to depth of a narrowest segment of each microchannel is from 0.5 to 0.01. Devices are further provided wherein a ratio of width to depth of a narrowest segment of each microchannel is about 0.05, 0.1, or 0.2. Devices are further provided wherein each microchannel of the plurality of microchannels has a total width of 30 um to 100 um. Devices are further provided wherein each microchannel of the plurality of microchannels has a total width of about 60 um. Devices are further provided wherein each microchannel of the plurality of microchannels has a depth of 10 to 500 um.
  • each microchannel of the plurality of microchannels has a depth of about 30 um.
  • the main channel has a width from 0.5 to 1.5 mm.
  • the main channel has a width of about 1.2 mm.
  • the main channel has a width of 1.15 mm.
  • the device comprises more than 250 main channels.
  • the device comprises more than 10,000 main channels.
  • Devices are further provided that further comprising a first molecule, wherein the first molecule is bound to an interior surface of the plurality of microchannels and comprises a reactive group that binds to a nucleoside phosphoramidite.
  • first molecule is a silane.
  • first molecule is an aminosilane.
  • first molecule is 11-acetoxyundecyltriethoxysilane, n-decyltriethoxysilane, (3-aminopropyl)trimethoxysilane, (3-aminopropyl)triethoxysilane, glycidyloxypropyl/trimethoxysilane or N-(3-triethoxysilylpropyl)-4-hydroxybutyramide.
  • Devices are further provided further comprising a second molecule, wherein the second molecule is bound to an interior surface of the main channel and lacks a reactive group that binds to a nucleoside phosphoramidite.
  • the second molecule is a fluorosilane.
  • the fluorosilane is (tridecafluorotetrahydrooctyl)-triethoxysilane.
  • the first molecule has a higher surface energy than the second molecule, and wherein the first molecule has a greater hydrophobicity than the second molecule.
  • the plate is a silicon plate.
  • Devices are further provided wherein the device comprises at least 30,000 microchannels. Devices are further provided wherein the device comprises at least 700,000 microchannels.
  • devices for synthesizing oligonucleic acids comprising: a plate; a main channel, wherein the main channel extends vertically into the silicon plate from an opening on a top side of the plate, and wherein the main channel has a width of 0.5 to 2 mm; and a plurality of microchannels connected to the main channel, wherein each microchannel of the plurality of microchannels extends vertically from an opening on a bottom side of the plate into the main channel, and wherein each microchannel of the plurality of microchannels comprises a total width that is less than 100 um, a microchannel surface area greater than 10,000 um, and a maximum width for the narrowest segment of the microchannel of 10 um.
  • each microchannel of the plurality of microchannels has a surface area greater than 12,000 um 2 .
  • each microchannel of the plurality of microchannels has a surface area of about 13,000 um 2 .
  • the plurality of microchannels comprises 50 to 500 microchannels.
  • the plurality of microchannels comprises 100 to 150 microchannels.
  • a ratio of width to depth of a narrowest segment of each microchannel is from 0.5 to 0.01.
  • Devices are further provided wherein a ratio of width to depth of a narrowest segment of each microchannel is about 0.05, 0.1, or 0.2.
  • each microchannel of the plurality of microchannels has a total width of 30 um to 100 um. Devices are further provided wherein each microchannel of the plurality of microchannels has a total width of about 60 um. Devices are further provided wherein each microchannel of the plurality of microchannels has a depth of 10 to 500 um. Devices are further provided wherein each microchannel of the plurality of microchannels has a depth of about 30 um. Devices are further provided wherein the main channel has a width from 0.5 to 1.5 mm. Devices are further provided wherein the main channel has a width of about 1.2 mm. Devices are further provided wherein the main channel has a width of 1.15 mm. Devices are further provided wherein the device comprises more than 250 main channels.
  • Devices are further provided wherein the device comprises more than 10,000 main channels.
  • Devices are further provided further comprising a first molecule, wherein the first molecule is bound to an interior surface of the plurality of microchannels and comprises a reactive group that binds to a nucleoside phosphoramidite.
  • Devices are further provided wherein the first molecule is a silane.
  • Devices are further provided wherein the first molecule is an aminosilane.
  • the first molecule is 11-acetoxyundecyltriethoxysilane, n-decyltriethoxysilane, (3-aminopropyl)trimethoxysilane, (3-aminopropyl)triethoxysilane, glycidyloxypropyl/trimethoxysilane or N-(3-triethoxysilylpropyl)-4-hydroxybutyramide.
  • Devices are further provided further comprising a second molecule, wherein the second molecule is bound to an interior surface of the main channel and lacks a reactive group that binds to a nucleoside phosphoramidite.
  • Devices are further provided wherein the second molecule is a fluorosilane. Devices are further provided wherein the fluorosilane is (tridecafluorotetrahydrooctyl)-triethoxysilane. Devices are further provided wherein the first molecule has a higher surface energy than the second molecule, and wherein the first molecule has a greater hydrophobicity than the second molecule. Devices are further provided wherein the plate is a silicon plate. Devices are further provided wherein the device comprises at least 30,000 microchannels. Devices are further provided wherein the device comprises at least 700,000 microchannels.
  • oligonucleic acid synthesis comprising: providing predetermined sequences for a plurality of non-identical oligonucleic acids; providing a device described herein; adding a droplet of fluid comprising an extension reaction reagent specific to a microchannel; allowing sufficient time for an extension reaction step to occur; and repeating steps (c) and (d) until the plurality of non-identical oligonucleic acids are synthesized, wherein each oligonucleic acid at least 10 bases in length and attached to an inside region of the microchannel, and wherein the synthesized non-identical oligonucleic acids encode sequences with an aggregate error rate of less than 1 in 2000 bases compared to the predetermined sequences.
  • Methods are further provided wherein the synthesized non-identical oligonucleic acids encode sequences with an aggregate error rate of less than 1 in 3000 bases compared to the predetermined sequences. Methods are further provided wherein the synthesized non-identical oligonucleic acids encode sequences with an insertion error rate of less than 1 in 5000 bases compared to the predetermined sequences. Methods are further provided wherein the synthesized non-identical oligonucleic acids encode sequences with a deletion error rate of less than 1 in 2000 bases compared to the predetermined sequences. Methods are further provided that further comprising washing the surface with a washing reagent, and wherein washing removes greater than 95% of unincorporated extension reaction reagent.
  • Methods are further provided wherein washing removes greater than 99% of unincorporated extension reaction reagent. Methods are further provided wherein the droplet of fluid is less than about 32 pL in volume. Methods are further provided wherein the method is completed in less than 24 hours. Methods are further provided wherein the synthesized plurality of non-identical oligonucleic acids are fluidically connected to a single main channel and collectively encode for a single gene. Methods are further provided wherein the oligonucleic acids collectively encode for at least 200 genes at least 1 kb in length.
  • Methods are further provided further comprising: releasing the plurality of non-identical oligonucleic acids from the surface; and subjecting the plurality of non-identical oligonucleic acids to a polymerase chain assembly reaction to assemble at least 200 genes.
  • Methods are further provided wherein the at least 200 genes have an aggregate error rate of less than 1 in 2000 bases compared to the predetermined sequences without correcting errors.
  • Methods are further provided wherein the at least 200 genes have an aggregate error rate of less than 1 in 3000 bases compared to the predetermined sequences without correcting errors.
  • each oligonucleic acid has a tether region 12 to 25 bases in length.
  • Methods are further provided wherein the tether region is homopolymeric.
  • Methods are further provided wherein each oligonucleic acid is at least 30 bases in length. Methods are further provided wherein each oligonucleic acid 50 to 500 bases in length.
  • devices for synthesizing oligonucleic acids comprising a silicon plate; a main channel, wherein the main channel extends vertically into the silicon plate from an opening on a top side of the silicon plate, and wherein the main channel has a width of 0.5 to 2 mm; and 50 to 500 microchannels connected to the main channel, wherein each of the 50 to 500 microchannels extends vertically from an opening on a bottom side of the silicon plate into the main channel, and wherein each microchannel of the 50 to 500 microchannels has a surface area to volume ratio of greater than about 0.4 (1/um), and a ratio of width to depth of a narrowest segment of each microchannel is from 0.5 to 0.01.
  • FIG. 1 illustrates a plate configured for oligonucleic acid synthesis comprising 24 regions, or sub-fields, each having an array of 256 clusters.
  • FIG. 2 illustrates a closer view of the sub-field in FIG. 1 having 16 ⁇ 16 of clusters, each cluster having 121 individual loci.
  • FIG. 3 illustrates a detailed view of the cluster in FIG. 2 , where the cluster has 121 loci.
  • FIG. 4 A illustrates a front view of a plate with a plurality of microchannels.
  • FIG. 4 B illustrates a sectional view of plate with a plurality of microchannels.
  • FIG. 5 illustrates three-dimensional arrangements for microchannels.
  • FIG. 6 illustrates a cluster having a plurality of loci with double comb shapes.
  • FIG. 7 illustrates a cluster having a plurality of loci with single comb shapes.
  • FIG. 8 illustrates a cluster having a plurality of loci with single serpentine shapes.
  • FIGS. 9 A- 9 F illustrate a workflow for the passive and active functionalization of an etched substrate.
  • FIGS. 10 A- 10 C illustrate reagent deposition directly into microchannels within a main channel, where: microchannels are actively functionalized ( FIG. 10 A ), main channels are passively functionalized ( FIG. 10 B ); and microchannels are actively functionalized and main channels are passively functionalized ( FIG. 10 C ).
  • the dotted lines indicate that the image depicts one main channel of many in a single substrate (e.g., a plate).
  • FIGS. 11 A- 11 C illustrate reagent deposition directly into microchannels within a main channel, where a plate contains a silicon oxide later at a boundary between a main channel and a microchannel, where: microchannels are actively functionalized ( FIG. 11 A ), main channels are passively functionalized ( FIG. 11 B ); and microchannels are actively functionalized and main channels are passively functionalized ( FIG. 11 C ).
  • the dotted lines indicate that the image depicts one main channel of many in a single substrate (e.g., a plate).
  • FIG. 12 illustrates a workflow for de novo oligonucleotide synthesis.
  • FIG. 13 illustrates a computer system
  • FIG. 14 is a block diagram illustrating architecture of a computer system.
  • FIG. 15 is a diagram demonstrating a network configured to incorporate a plurality of computer systems, a plurality of cell phones and personal data assistants, and Network Attached Storage (NAS).
  • NAS Network Attached Storage
  • FIG. 16 is a block diagram of a multiprocessor computer system using a shared virtual address memory space.
  • FIG. 17 depicts read counts from a sub-array having 256 clusters (left), and an image of a cluster having 121 loci (right).
  • FIG. 18 is a graphical representation of oligonucleic acid frequency versus abundance from an experiment where oligonucleic acids were synthesized on the substrate of FIG. 17 .
  • FIG. 19 is a graphical representation of oligonucleic acid frequency versus abundance for four representative clusters of the substrate of FIG. 17 .
  • FIG. 20 is a graphical representation of oligonucleic acid frequency versus error rate for oligonucleic acids synthesized on the substrate of FIG. 17 .
  • FIG. 21 is a graphical representation of oligonucleic acid frequency versus error rate for oligonucleic acids synthesized on four representative clusters of the substrate of FIG. 17 .
  • FIG. 22 is a representation of read counts from 240 assembled genes from a library of oligonucleic acids synthesized on a substrate.
  • FIGS. 23 , 24 , 25 and 26 provide digital images from gel electrophoresis of 240 assembled genes from a library of oligonucleic acids synthesized on the substrate of FIG. 17 .
  • FIG. 27 provides an output reading from next generation sequencing of 240 assembled genes from a library of oligonucleic acids synthesized on the substrate of FIG. 17 .
  • FIG. 28 provides a graphical representation of insertion/deletion (“indel”) error count as a function of read cycle for a synthesized library of oligonucleic acids.
  • FIG. 29 provides a digital image of an electrophoresis gel showing 25mer to 200mer oligonucleic acids synthesized using a substrate and methods provided herein.
  • the present disclosure provides compositions, devices, methods and systems for the de novo synthesis of a library of oligonucleic acids with low error rates.
  • the oligonucleic acids are useful components, such as for the generation of larger nucleic acids, such as genes as part of gene libraries.
  • microchannels small channels which also serve as locations for oligonucleic acid extension.
  • Factors that can impact the flow of fluid throw the surface include, without limitation, the number of microchannels, microchannel size, the shape of the microchannels, the width of a main channel which a group of microchannels collectively connect, and the chemical properties of surfaces involved (e.g., hydrophobicity and surface energy).
  • Rapid fluid transfer is desirable to provide for efficient chemical exchange during various steps of the de novo nucleic acid synthesis process, and reduce unwanted side reactions that may lead to increased error rates.
  • Devices described herein increase fluid transfer rate through a substrate and also increase the amount of surface available for nucleic acid extension is by having microchannels with a high surface area to volume ratio. Such devices also provide for synthesis of oligonucleic acids with low error rates.
  • oligonucleic acids synthesized within a cluster of extension locations comprise specific predetermined sequences that are configured to be assembled to generate a larger nucleic acid. In this manner, the parallel generation of genes is done on a single substrate.
  • the average error rates for oligonucleic acids synthesized within a library using the systems and methods provided are often less than 1 in 1000, and are preferably less than about 1 in 2000, 1 in 5000 or less often.
  • locus refers to a discrete region on a structure which provides support for extension of an oligonucleic acid.
  • preselected sequence As used herein, the terms “preselected sequence,” “predefined sequence” or “predetermined sequence” are used interchangeably. The terms refer to sequence of a polymer that is known and chosen before synthesis or assembly of the polymer. In particular, various aspects of the invention are described herein primarily with regard to the preparation of nucleic acid molecules, the sequence of the oligonucleotide or polynucleotide being known and chosen before the synthesis or assembly of the nucleic acid molecules.
  • FIG. 1 An exemplary substrate 100 is illustrated in FIG. 1 , wherein the substrate 100 has about the same size dimensions as a standard 96 well plate: 140 mm by 90 mm.
  • the substrate 100 comprises clusters grouped in 24 regions or sub-fields 105 , each sub-field 105 comprising an array of 256 clusters 110 .
  • FIG. 2 An expanded view of an exemplary sub-field 105 is shown in FIG. 2 . In the expanded view of four clusters ( FIG.
  • a single cluster 110 has a Y axis cluster pitch (distance from center to center of adjacent clusters) of 1079.210 um or 1142.694 um, and an X axis cluster pitch of 1125 um.
  • An illustrative cluster 110 is depicted in FIG. 3 , where the Y axis loci pitch (distance from center to center of adjacent loci) is 63.483 um, and an X axis loci pitch is 75 um.
  • the locus width at the longest part e.g., diameter for a circular loci, is 50 um and the distance between loci is 24 um.
  • the number of loci 305 in the exemplary cluster in FIG. 3 is 121.
  • FIGS. 4 A- 4 B An exemplary arrangement is illustrated in FIGS. 4 A- 4 B where a plate 405 is illustrated comprising a main channel 410 and a plurality of microchannels 415 connected to the main channel 410 .
  • the connection between the main channel 410 and the plurality of microchannels 415 provides for a fluid communication for flow paths from the main channel 410 to the each of the plurality of microchannels 415 .
  • a plate 405 described herein can comprise multiple main channels 410 .
  • the plurality of microchannels 415 collectively forms a cluster within the main channel 410 .
  • a library of oligonucleic acids is synthesized in a plurality of loci where the loci are collectively a plurality of microchannels 415 of a cluster where the cluster is within a main channel 410 , followed by the assembly of the oligonucleic acids into a large nucleic acid such as gene, wherein the assembly of the large nucleic acid optionally occurs within a main channel of the cluster, e.g., by using PCA.
  • a different oligonucleic acid is grown in each of the microchannels 415 with a main channel 410 , and the oligonucleic acids collectively encode for a single gene.
  • the structure is configured to allow for controlled flow for de novo oligonucleic acid synthesis by providing for rapid exchange of chemical exposure during de novo synthesis of oligonucleic acids.
  • configuration described herein provide for the controlled and even distribution of mass transfer paths, chemical exposure times, and/or wash efficiency during oligonucleic acid synthesis.
  • the configuration of a substrate allows for increased sweep efficiency, for example by providing sufficient volume for a growing an oligonucleic acid such that the excluded volume by the growing oligonucleic acid does not take up more than 50, 45, 40, 35, 30, 25, 20, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2, 1%, or less of the initially available volume that is available or suitable for growing the oligonucleic acid.
  • chemical coatings also provide and additional means for segregating oligonucleic acid species situated within a microchannel. Segregation can be achieved by differential functionalization of the surface, for example by having active and passive regions for oligonucleic acid synthesis coated on the surface. Differential functionalization can also be achieved by alternating the hydrophobicity across the substrate surface, thereby creating water contact angle effects that may cause beading or wetting of the deposited reagents. Employing larger structures can decrease splashing and cross-contamination of distinct oligonucleic acid synthesis locations with reagents of the neighboring spots. A device, such as an oligonucleic acid synthesizer, may be used to deposit reagents to distinct oligonucleic acid synthesis locations.
  • Microchannels Structural Features
  • Microchannels described herein provide chemical properties, dimensions (width, height/depth, or length) de novo synthesized oligonucleic acids having a low error rate. While the plurality of microchannels 415 in FIG. 4 are circular, various shapes can be used to enhance flow rates through the channel, e.g., microchannels with curves or combs. A microchannel having a shape providing an increased surface area to volume ratio may be desirable for several reasons. First, an increase in surface area provides an increase in area that is suitable for oligonucleic acid attachment and synthesis. Second, a locus in the shape of a microchannel with increased surface area to volume ratio requires less fluid for efficient flow through the channel, thereby allowing less reagent volume per reaction.
  • the efficient flow through a locus in the shape of a microchannel with increased surface area to volume ratio minimizes residual occupation of reagents during flow through the microchannel and enhances wash efficiency, thereby minimizing or essentially eliminating undesirable secondary reactions during the chemical steps involved in the oligonucleotide extension process. Minimizing undesirable secondary reactions is another factor for keeping error rate down during oligonucleic acid synthesis.
  • FIG. 5 shows a top view of a double comb 510 , single comb 520 or serpentine microchannel shape 530 .
  • the width of the microchannel at the narrowest segment is 5 um, and each of the microchannel comprises at least one turn greater than 90 degrees in total.
  • the microchannel comprises 1 to 10 or more turns greater than 90 degrees in total.
  • the turns are curved and the microchannel comprises 1 to 10 or more turns in total.
  • the turns are 90 degrees, i.e. perpendicular fluid paths from a top view.
  • the turns are curved and 180 degrees, i.e. a U turn is viewed from top view.
  • a turn in a microchannel fluid path is 45 to 180 degrees in total, when viewed from a top view.
  • a main channel 600 comprises a cluster of double comb channels, where each double comb channel 510 has a total width of 57 um, the width of the microchannel at the narrowest segment is 5 um, and each stick of the “comb” is 14 um apart from the center of another stick ( FIG. 6 ).
  • a main channel 700 comprises a cluster of single comb channels, where each single comb channel 520 has a total width of 49 um, the width of the microchannel at the narrowest segment is 5 um, and each stick of the “comb” is 14 um apart from the center of another stick ( FIG. 7 ).
  • a main channel 800 comprises a cluster of serpentine shaped channels, where each serpentine shaped channel 530 has a total width of 54 um, the width of the microchannel at the narrowest segment is 5 um, and each turn of the shape results in a parallel region 14 um apart from another region ( FIG. 7 ).
  • the microchannel extends vertically into the substrate, e.g., a plate.
  • a total surface area for a locus (e.g., a microchannel) in a device described herein is greater than about 9000, 10000, 11000, 12000, 12500, 12600, 12700, 12800, 12900 or 13000 um 2 . In some instances, the total surface area for a locus in a device described herein is about 10000 to about 15000 um 2 . In some instances, the total surface area for a locus in a device described herein is about 12000 to about 13000 um 2 . In some instances, the total surface area to volume ratio (1/um) for a locus in a device described herein is about 0.2 to 0.5.
  • the total surface area to volume ratio (1/um) for a locus in a device described herein is greater than 0.20. In some instances, the total surface area to volume ratio (1/um) for a locus in a device described herein is greater than about 0.40. In some instances, the total surface area to volume ratio (1/um) for a locus in a device described herein is about 0.40. In some instances, the total surface area to volume ratio (1/um) for a locus in a device described herein is 0.41.
  • a microchannel described herein has a width to depth (or height) ratio of 1 to 0.01, wherein the width is a measurement of the width at the narrowest segment of the microchannel. In some instances, a microchannel described herein has a width to depth (or height) ratio of 0.5 to 0.01, wherein the width is a measurement of the width at the narrowest segment of the microchannel. In some instances, a microchannel described herein has a width to depth (or height) ratio of about 0.01, 0.05, 0.1, 0.15, 0.16, 0.2, 0.5, or 1.
  • a substrate comprises a plurality of microchannels corresponding to a plurality of loci within a cluster, wherein the height or depth of the microchannel is from about 5 um to about 500 um, from about 5 um to about 400 um, from about 5 um to about 300 um, from about 5 um to about 200 um, from about 5 um to about 100 um, from about 5 um to about 50 um, or from about 10 um to about 50 um.
  • the height of a microchannel is less than 100 um, less than 80 um, less than 60 um, less than 40 um or less than 20 um.
  • microchannel height is about 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 200, 300, 400, 500 ⁇ m or more.
  • the width of a locus is from about 0.5 ⁇ m to about 500 um, from about 1 um to about 200 um, from about 1 um to about 100 um, from about 5 um to about 100 um, or from about 0.1 um to about 100 um, for example, about 90 um, 80 um, 70 um, 60 um, 50 um, 40 um, 30 um, 20 um, 10 um, 5 um, 1 um or 0.5 um.
  • the width of a locus is less than about 100 um, 90 um, 80 um, 70 um, 60 um, 50 um, 40 um, 30 um, 20 um or 10 um.
  • the distance between the center of two adjacent loci is from about 1 um to about 500 um, from about 1 um to about 200 um, from about 1 um to about 100 um, from about 5 um to about 200 um, from about 5 um to about 100 um, from about 5 um to about 50 um, or from about 5 um to about 30 um, for example, about 20 um.
  • the total width of a microchannel is about 10 um, 20 um, 30 um, 40 um, 50 um, 60 um, 70 um, 80 um, 90 um, or 100 um. In some instances, the total width of a microchannel is about 10 um to 100 um, 30 um to 100 um, or 50 um to 70 um.
  • each locus supports the synthesis of a population of oligonucleic acids having a different sequence than a population of oligonucleic acids grown on another locus.
  • surfaces which comprise at least 10, 100, 256, 500, 1000, 2000, 3000, 4000, 5000, 6000, 7000, 8000, 9000, 10000, 11000, 12000, 13000, 14000, 15000, 20000, 30000, 40000, 50000 or more clusters.
  • each cluster includes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 120, 130, 150, 200, 500 or more loci.
  • each cluster includes 50 to 500, 50 to 200, 50 to 150, or 100 to 150 loci.
  • each cluster includes 100 to 150 loci.
  • each cluster includes 109, 121, 130 or 137 loci.
  • the distance between the centers of two adjacent loci within a cluster is from about 1 um to about 500 um, from about 5 um to about 200 um, or from about 0.5 ⁇ m to about 100 um.
  • the distance between two centers of adjacent loci is about 0.5 ⁇ m, 20 um, 25 um, 30 um, 40 um, 50 um, 60 um, 70 um, 80 um, 90 um, or 100 um.
  • loci having a width at the longest segment of 5 to 100 um. In some cases, the loci have a width at the longest segment of about 30, 35, 40, 45, 50, 55 or 60 um. In some cases, the loci are microchannels having multiple segments, wherein each segment has a center to center distance apart of 5 to 50 um. In some cases, the center to center distance apart for each segment is about 5, 10, 15, 20 or 25 um.
  • Main channels described herein extend from a top surface of a substrate, e.g., a plate, into the plate until reaching an interface with a plurality of microchannels, each microchannel connecting to a bottom surface of the substrate. In some instances, the main channel extends vertically.
  • Main channels e.g., main channel 410 , can be in circular, rectangular, tapered, or rounded shapes.
  • a width (a diameter in the case of a circle) of a cluster or the width of a main channel comprising a cluster, or both is between about 0.05 mm to about 50 mm, between about 0.05 mm to about 10 mm, between about 0.05 mm and about 5 mm, between about 0.05 mm and about 4 mm, between about 0.05 mm and about 3 mm, between about 0.05 mm and about 2 mm, between about 0.05 mm and about 1 mm, between about 0.05 mm and about 0.5 mm, between about 0.05 mm and about 0.1 mm, between about 0.1 mm and 10 mm, between about 0.2 mm and 10 mm, between about 0.3 mm and about 10 mm, between about 0.4 mm and about 10 mm, between about 0.5 mm and 10 mm, between about 0.5 mm and about 5 mm, between about 0.5 mm and about 1.5 mm, or between about 0.5 mm and about 2 mm.
  • the width of a cluster or main channel or both is less than or about 5 mm, 4 mm, 3 mm, 2 mm, 1.5 mm, 1.2 mm, 1.15 mm, 1 mm, 0.5 mm, 0.1 mm, 0.09 mm, 0.08 mm, 0.07 mm, 0.06 mm or 0.05 mm. In some instances, the width of a cluster or main channel is between about 1.0 and about 1.3 mm. In some instances, the width of a cluster or main channel, or both is about 1.150 mm. In some instances, the width of a cluster or main channel, or both is about 0.08 mm.
  • the height (depth) of a main channel is from about 20 um to about 1000 um, from about 50 um to about 1000 um, from about 100 ⁇ m to about 1000 um, from about 200 ⁇ m to about 1000 um, from about 300 ⁇ m to about 1000 um, from about 400 um to about 1000 um, or from about 500 ⁇ m to about 1000 um. In some cases, the height of a main channel is less than about 1000 um, less than about 900 um, less than about 800 um, less than about 700 um, or less than about 600 um. In some cases, the height of a main channel is about 500 um. In some cases, the height of a main channel is about 450 um.
  • the number of distinct nucleic acids or genes assembled from a plurality of oligonucleic acids synthesized on a substrate is dependent on the number of clusters available in the substrate.
  • the density of clusters within a substrate is at least or about 1 cluster per 100 mm 2 , 1 cluster per 10 mm 2 , 1 cluster per 5 mm 2 , 1 cluster per 4 mm 2 , 1 cluster per 3 mm 2 , 1 cluster per 2 mm 2 , 1 cluster per 1 mm 2 , 2 clusters per 1 mm 2 , 3 clusters per 1 mm 2 , 4 clusters per 1 mm 2 , 5 clusters per 1 mm 2 , 10 clusters per 1 mm 2 , 50 clusters per 1 mm 2 or more.
  • a substrate comprises from about 1 cluster per 10 mm 2 to about 10 clusters per 1 mm 2 .
  • the distance between the centers of two adjacent clusters is less than about 50 um, 100 um, 200 um, 500 um, 1000 um, or 2000 um or 5000 um. In some cases, the distance between the centers of two adjacent clusters is between about 50 um and about 100 um, between about 50 um and about 200 um, between about 50 um and about 300 um, between about 50 um and about 500 um, and between about 100 ⁇ m to about 2000 um.
  • the distance between the centers of two adjacent clusters is between about 0.05 mm to about 50 mm, between about 0.05 mm to about 10 mm, between about 0.05 mm and about 5 mm, between about 0.05 mm and about 4 mm, between about 0.05 mm and about 3 mm, between about 0.05 mm and about 2 mm, between about 0.1 mm and 10 mm, between about 0.2 mm and 10 mm, between about 0.3 mm and about 10 mm, between about 0.4 mm and about 10 mm, between about 0.5 mm and 10 mm, between about 0.5 mm and about 5 mm, or between about 0.5 mm and about 2 mm.
  • the number of distinct oligonucleic acids synthesized on a substrate is dependent on the number of distinct loci available in the substrate.
  • the density of loci within a cluster of a substrate is at least or about 1 locus per mm 2 , 10 loci per mm 2 , 25 loci per mm 2 , 50 loci per mm 2 , 65 loci per mm 2 , 75 loci per mm 2 , 100 loci per mm 2 , 130 loci per mm 2 , 150 loci per mm 2 , 175 loci per mm 2 , 200 loci per mm 2 , 300 loci per mm 2 , 400 loci per mm 2 , 500 loci per mm 2 , 1,000 loci per mm 2 or more.
  • a substrate comprises from about 10 loci per mm 2 to about 500 mm 2 , from about 25 loci per mm 2 to about 400 mm 2 , from about 50 loci per mm 2 to about 500 mm 2 , from about 100 loci per mm 2 to about 500 mm 2 , from about 150 loci per mm 2 to about 500 mm 2 , from about 10 loci per mm 2 to about 250 mm 2 , from about 50 loci per mm 2 to about 250 mm 2 , from about 10 loci per mm 2 to about 200 mm 2 , or from about 50 loci per mm 2 to about 200 mm 2 .
  • the distance between the centers of two adjacent loci within a cluster is from about 10 um to about 500 um, from about 10 um to about 200 um, or from about 10 um to about 100 um. In some cases, the distance between two centers of adjacent loci is greater than about 10 um, 20 um, 30 um, 40 um, 50 um, 60 um, 70 um, 80 um, 90 um or 100 um. In some cases, the distance between the centers of two adjacent loci is less than about 200 um, 150 um, 100 um, 80 um, 70 um, 60 um, 50 um, 40 um, 30 um, 20 um or 10 um.
  • a device described herein may comprise multiple main channels.
  • a device described herein comprises 1 to 250, 2 to 250, 1 to 500 or more main channels.
  • a device described herein comprises about 2, 10, 50, 100, 150, 200, 250, 256, 500, 512, 1000, 2500, 3000, 4000, 5000, 6000, 6144, 10000 or more main channels.
  • a plate described herein comprises about 2, 10, 50, 100, 150, 200, 250, 256, 500, 512, 1000, 2500, 3000, 4000, 5000, 6000, 6144, 10000 or more main channels.
  • the plate is a silicon plate or a silicon on insulator (SOI) plate.
  • a substrate comprises a surface that supports the synthesis of a plurality of oligonucleic acids having different predetermined sequences at addressable locations on a common support.
  • a substrate described herein provides support for the synthesis of more than 2,000; 5,000; 10,000; 20,000; 30,000; 50,000; 100,000; 200,000; 300,000; 400,000; 500,000; 600,000; 700,000; 800,000; 900,000; 1,000,000; 1,200,000; 1,400,000; 1,600,000; 1,800,000; 2,000,000; 2,500,000; 3,000,000; 3,500,000; 4,000,000; 4,500,000; 5,000,000; 10,000,000 or more non-identical oligonucleic acids.
  • the substrate provides support for the synthesis of more than 2,000; 5,000; 10,000; 20,000; 50,000; 100,000; 200,000; 300,000; 400,000; 500,000; 600,000; 700,000; 800,000; 900,000; 1,000,000; 1,200,000; 1,400,000; 1,600,000; 1,800,000; 2,000,000; 2,500,000; 3,000,000; 3,500,000; 4,000,000; 4,500,000; 5,000,000; 10,000,000 or more oligonucleic acids encoding for distinct sequences.
  • at least a portion of the oligonucleic acids have an identical sequence or are configured to be synthesized with an identical sequence.
  • the substrate provides a surface environment for the growth of oligonucleic acids having at least about 50, 60, 70, 75, 80, 85, 90, 95, 100, 110, 120, 130, 140, 150, 160, 175, 200, 225, 250, 275, 300, 325, 350, 375, 400, 425, 450, 475, 500 bases or more.
  • oligonucleic acids are synthesized on distinct loci of a substrate, wherein each locus supports the synthesis of a population of oligonucleic acids. In some cases, each locus supports the synthesis of a population of oligonucleic acids having a different sequence than a population of oligonucleic acids grown on another locus. In some instances, the loci of a substrate are located within a plurality of clusters. In some instances, a substrate comprises at least 10, 500, 1000, 2000, 3000, 4000, 5000, 6000, 7000, 8000, 9000, 10000, 11000, 12000, 13000, 14000, 15000, 20000, 30000, 40000, 50000 or more clusters.
  • a substrate comprises more than 2,000; 5,000; 10,000; 100,000; 200,000; 300,000; 400,000; 500,000; 600,000; 700,000; 800,000; 900,000; 1,000,000; 1,100,000; 1,200,000; 1,300,000; 1,400,000; 1,500,000; 1,600,000; 1,700,000; 1,800,000; 1,900,000; 2,000,000; 300,000; 400,000; 500,000; 600,000; 700,000; 800,000; 900,000; 1,000,000; 1,200,000; 1,400,000; 1,600,000; 1,800,000; 2,000,000; 2,500,000; 3,000,000; 3,500,000; 4,000,000; 4,500,000; 5,000,000; or 10,000,000 or more distinct loci.
  • each cluster includes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 120, 130, 150 or more loci. In some instances, each cluster includes 50 to 500, 100 to 150, or 100 to 200 loci. In some instances, each cluster includes 109, 121, 130 or 137 loci. In some instances, each cluster includes 5, 6, 7, 8, 9, 10, 11 or 12 loci.
  • oligonucleic acids from distinct loci within one cluster have sequences that, when assembled, encode for a contiguous longer oligonucleic acid of a predetermined sequence, for example, a gene.
  • a substrate comprising more than 20,000 loci e.g., microchannels
  • a substrate comprising more than 29,000 loci e.g., microchannels
  • a substrate comprising more than 700,000 loci is used for the synthesis of about 6,000 distinct genes of predetermined sequence.
  • Substrates provided may be fabricated from a variety of materials suitable for the methods and compositions described herein.
  • substrate materials are fabricated to exhibit a low level of nucleotide binding.
  • substrate materials are modified to generate distinct surfaces that exhibit a high level of nucleotide binding.
  • substrate materials are transparent to visible and/or UV light.
  • substrate materials are sufficiently conductive, e.g., are able to form uniform electric fields across all or a portion of a substrate.
  • conductive materials may be connected to an electric ground.
  • the substrate is heat conductive or insulated.
  • the materials are chemical resistant and heat resistant to support chemical or biochemical reactions, for example oligonucleic acid synthesis reaction processes.
  • a substrate comprises flexible materials.
  • Flexible materials include, without limitation, modified nylon, unmodified nylon, nitrocellulose, polypropylene, and the like.
  • a substrate comprises rigid materials. Rigid materials include, without limitation, glass, fuse silica, silicon, silicon dioxide, silicon nitride, plastics (for example, polytetraflouroethylene, polypropylene, polystyrene, polycarbonate, and blends thereof, and the like), and metals (for example, gold, platinum, and the like).
  • a substrate is fabricated from a material comprising silicon, polystyrene, agarose, dextran, cellulosic polymers, polyacrylamides, polydimethylsiloxane (PDMS), glass, or any combination thereof.
  • the substrates may be manufactured with a combination of materials listed herein or any other suitable material known in the art.
  • substrates described herein are in the shape of a plate, film or tape.
  • a substrate is about the size of a standard 96 well plate, for example between about 100 and 200 mm by between about 50 and 150 mm.
  • a substrate has a diameter less than or equal to about 1000 mm, 500 mm, 450 mm, 400 mm, 300 mm, 250 nm, 200 mm, 150 mm, 100 mm or 50 mm.
  • the diameter of a substrate is between about 25 mm and 1000 mm, between about 25 mm and about 800 mm, between about 25 mm and about 600 mm, between about 25 mm and about 500 mm, between about 25 mm and about 400 mm, between about 25 mm and about 300 mm, or between about 25 mm and about 200.
  • Non-limiting examples of substrate size include about 300 mm, 200 mm, 150 mm, 130 mm, 100 mm, 76 mm, 51 mm and 25 mm.
  • a substrate has a planar surface area of at least about 100 mm 2 ; 200 mm 2 ; 500 mm 2 ; 1,000 mm 2 ; 2,000 mm 2 ; 5,000 mm 2 ; 10,000 mm 2 ; 12,000 mm 2 ; 15,000 mm 2 ; 20,000 mm 2 ; 30,000 mm 2 ; 40,000 mm 2 ; 50,000 mm 2 or more.
  • the thickness of a substrate is between about 50 mm and about 2000 mm, between about 50 mm and about 1000 mm, between about 100 mm and about 1000 mm, between about 200 mm and about 1000 mm, or between about 250 mm and about 1000 mm.
  • substrate thickness include 275 mm, 375 mm, 525 mm, 625 mm, 675 mm, 725 mm, 775 mm and 925 mm.
  • the thickness of a substrate varies with diameter and depends on the composition of the substrate. For example, a substrate comprising materials other than silicon may have a different thickness than a silicon substrate of the same diameter. Substrate thickness may be determined by the mechanical strength of the material used and the substrate must be thick enough to support its own weight without cracking during handling.
  • a substrate described herein comprises a plurality of smaller regions, for example, at least about 2, 4, 6, 8, 10, 16, 24, 39, 50, 100 or more regions, wherein each region may be used independently from another region.
  • regions of a substrate are sub-fields or chips of a substrate.
  • reference to a substrate includes a region of a substrate.
  • Selective deposition or selective functionalization refers to a process that produces two or more distinct areas on a structure, wherein at least one area has a different surface or chemical property that another area of the same structure. Such properties include, without limitation, surface energy, chemical termination, surface concentration of a chemical moiety, and the like.
  • active functionalization refers to a method comprising the functionalization of a surface that will be utilized for oligonucleic acid synthesis.
  • passive functionalization refers to a method comprising the functionalization of a surface that will render these areas ineffective for oligonucleic acid synthesis.
  • any suitable process that changes the chemical properties of the surface described herein or known in the art may be used to functionalize the surface, for example chemical vapor deposition of an organosilane. Typically, this results in the deposition of a self-assembled monolayer (SAM) of the functionalization species.
  • SAM self-assembled monolayer
  • a method for functionalizing a surface of a substrate for oligonucleic acid synthesis comprises a resist or photoresist coat.
  • Photoresist in many cases, refers to a light-sensitive material useful in photolithography to form patterned coatings. It can be applied as a liquid to solidify on a substrate as volatile solvents in the mixture evaporate.
  • the resist is applied in a spin coating process as a thin film, e.g., 1 um to 100 um.
  • the coated resist is patterned by exposing it to light through a mask or reticle, changing its dissolution rate in a developer.
  • the resist cost is used as a sacrificial layer that serves as a blocking layer for subsequent steps that modify the underlying surface, e.g., etching, and then is removed by resist stripping.
  • the flow of resist throughout various features of the structure is controlled by the design of the structure.
  • a surface of a structure is functionalized while areas covered in resist are protected from active or passive functionalization.
  • a substrate suitable for functionalization e.g., an etched substrate comprising three-dimensional features
  • resist for example, by an oligonucleic acid synthesizer devices capable of delivering drops of fluid with micrometer accuracy.
  • a droplet of resist may be deposited into the lowered feature where it optionally spreads.
  • a portion of the resist is removed, for example, by etching (e.g., oxygen plasma etch) to leave a smooth surface covering only a select area.
  • the substrate is passively functionalized with a passive functionalization agent comprising a chemically inert moiety to create a surface having low surface energy.
  • the passively functionalized substrate is resist stripped, exposing areas of the substrate that were exposed during front-end processing (etched regions).
  • FIGS. 9 A- 9 F depicts a workflow for active functionalization of a microchannels and passive functionalization of surrounding areas, e.g., main channels.
  • An etched substrate 905 is prepared for active functionalization.
  • the substrate is wet cleaned, for example, using a piranha solution.
  • the substrate is plasma cleaned, for example, by dry oxygen plasma exposure.
  • the device layer is coated with photoresist 910 , optionally after cleaning.
  • the photoresist is coated by a process governed by wicking into the device layer channels.
  • the photoresist is patterned using photolithography to expose areas that are desired to be passive (i.e., areas where oligonucleic acid synthesis is not designed to take place). Patterning by photolithography may occur by exposing the resist to light through a binary mask that has a pattern of interest. After exposure, the resist in the exposed regions may be removed in developer solution in FIG. 9 C , and leaves photoresist at predetermined regions 915 .
  • a passive functionalization agent 920 such as a low surface energy silane (e.g., fluorosilane gas vapor), for example, by chemical vapor deposition (CVD). Exposure to fluorosilane gas results in the deposition of a fluorocarbon on the surfaces without photoresist.
  • the substrate is exposed to a hydrocarbon silane.
  • passive functionalization comprises exposing a substrate to a passive functionalization agent such as one comprising silane.
  • the passively functionalized substrate are unresponsive to additional layers of functionalization agent (e.g., active functionalization agent) creating a monolayer on the surface.
  • Resist stripping as shown in FIG. 9 E leaves some surfaces passively functionalized while exposing regions of the substrate that was underneath the resist 925 .
  • a resist is dissolved in an organic solvent.
  • resist stripping of a substrate that was passively functionalized with a fluorosilane gas leaves a surface having some regions of fluorination. In some cases, regions that were underneath the resist comprise silicon or silicon dioxide.
  • the surface is actively functionalized to prepare the surface for oligonucleic acid synthesis.
  • An exemplary active functionalization agent is one that has a higher surface energy than the passive functionalization agent.
  • a substrate is first cleaned, for example, using a piranha solution.
  • a cleaning process includes soaking a substrate in a piranha solution (e.g., 90% H 2 SO 4 , 10% H 2 O 2 ) at an elevated temperature (e.g., 120° C.) and washing (e.g., water) and drying the substrate (e.g., nitrogen gas).
  • the process optionally includes a post piranha treatment comprising soaking the piranha treated substrate in a basic solution (e.g., NH 4 OH) followed by an aqueous wash (e.g., water).
  • a substrate is plasma cleaned, optionally following the piranha soak and optional post piranha treatment.
  • An example of a plasma cleaning process comprises an oxygen plasma etch.
  • the surface is deposited with an active functionalization agent following by vaporization.
  • the substrate is actively functionalized prior to cleaning, for example, by piranha treatment and/or plasma cleaning.
  • the process for substrate functionalization optionally comprises a resist coat and a resist strip.
  • the substrate is spin coated with a resist, for example, SPRTM 3612 positive photoresist.
  • the process for substrate functionalization in various instances, comprises lithography with patterned functionalization. In some instances, photolithography is performed following resist coating. In some instances, after lithography, the substrate is visually inspected for lithography defects.
  • the process for substrate functionalization in some instances, comprises a cleaning step, whereby residues of the substrate are removed, for example, by plasma cleaning or etching. In some instances, the plasma cleaning step is performed at some step after the lithography step.
  • a substrate coated with a resist is treated to remove the resist, for example, after functionalization and/or after lithography.
  • the resist is removed with a solvent, for example, with a stripping solution comprising N-methyl-2-pyrrolidone.
  • resist stripping comprises sonication or ultrasonication.
  • a resist is coated and stripped, followed by active functionalization of the exposed areas to create a desired differential functionalization pattern.
  • the methods and compositions described herein relate to the application of photoresist for the generation of modified surface properties in selective areas, wherein the application of the photoresist relies on the fluidic properties of the substrates defining the spatial distribution of the photoresist.
  • surface tension effects related to the applied fluid may define the flow of the photoresist.
  • surface tension and/or capillary action effects may facilitate drawing of the photoresist into small structures in a controlled fashion before the resist solvents evaporate.
  • resist contact points are pinned by sharp edges, thereby controlling the advance of the fluid.
  • the underlying structures may be designed based on the desired flow patterns that are used to apply photoresist during the manufacturing and functionalization processes.
  • a solid organic layer left behind after solvents evaporate may be used to pursue the subsequent steps of the manufacturing process.
  • Substrates may be designed to control the flow of fluids by facilitating or inhibiting wicking effects into neighboring fluidic paths.
  • a substrate is designed to avoid overlap between top and bottom edges, which facilitates the keeping of the fluid in top structures allowing for a particular disposition of the resist.
  • the top and bottom edges overlap, leading to the wicking of the applied fluid into bottom structures. Appropriate designs may be selected accordingly, depending on the desired application of the resist.
  • FIG. 10 A illustrates the deposition of reagents 1005 on a plate 405 .
  • the substrate 405 comprises a plurality of microchannels 415 in fluidic connection with a main channel 410 .
  • a single main channel 410 is depicted, and the dashed lines indicate that this is just one of many main channel 410 /plurality of microchannels 415 connections in a single plate 405 .
  • Each of the plurality of microchannels 415 is coated with an active functionalization agent 1015 .
  • Each microchannel 415 of the plurality of microchannels in the example has a width of 5 um and a depth of 30 um.
  • the main channel 410 has a diameter of 1150 um.
  • the total height of the plate is 450 um.
  • the main channel 410 encircles a cluster of microchannels.
  • a substrate described herein is coated with a layer of material comprising on or more passive functionalization agents.
  • a passive functionalization agent 1020 coats surface of a plate 405 .
  • a substrate described herein is coated with a layer of material comprising on or more passive functionalization agents and the microchannels of the substrate are coated with one or more active functionalization agents.
  • the starting substrate is a silicon on insulator plate, where layers of silicon sandwich an insulator layer 1110 , typically silicon dioxide.
  • the thickness of the insulator layer 1110 is from 1 to 50 um, e.g., 20 um.
  • an active functionalization agent is a molecule that binds to the surface of the substrate and is also capable of binding to a nucleic acid monomer, thereby supporting a coupling reaction to the surface.
  • Exemplary active functionalization agents are molecules having a hydroxyl group available for coupling with a nucleoside in a coupling reaction.
  • only main channels and/or surrounding areas (and not the microchannels) in a device described herein are coated with passive functionalization agent.
  • a passive functionalization agent is a molecule that binds to the surface of the substrate and lacks a moiety available for coupling with a nucleoside in a coupling reaction.
  • Oligonucleic acids synthesized in the channels may be released for the purposes of generating longer nucleic acids.
  • oligonucleic acids within one cluster are released from their respective surfaces and pool into the main channel.
  • the pooled oligonucleic acids are assembled into a larger nucleic acid, such as a gene, within the main channel, so that the main channel functions as a reactor for nucleic acid assembly.
  • oligonucleic acids within one cluster are released from their respective surfaces and pool into a nanoreactor in fluidic communication with the microchannels.
  • nucleic acid verification e.g., sequencing of oligonucleic acids and/or assembled genes
  • Nucleic acid assembly includes polymerase cycling assembly (PCA).
  • PCA polymerase cycling assembly
  • a capping element or other device is placed over open sides of the main channel to create an enclosed reactor.
  • a substrate comprising a main channel that functions as a reactor for each cluster has the advantage that each cluster may have a different environment from another cluster in another reactor.
  • sealed reactors e.g., those with capping elements
  • a substrate is configured for both active and passive functionalization moieties bound to the surface at different areas of the substrate surface, generating distinct regions for oligonucleic acid synthesis to take place.
  • both active and passive functionalization agents are mixed within a particular region of the surface. Such a mixture provides a diluted region of active functionalization agent and therefore lowers the density of functionalization agent in a particular region.
  • Substrates described herein may comprise a high surface energy region at the site of active functionalization agent deposition.
  • the high surface energy region is coated with aminosilane.
  • the silane group binds to the surface, while the rest of the molecule provides a distance from the surface and a free group at the end to which incoming bases attach.
  • the free group is a hydroxyl group.
  • the high surface energy region includes an active functionalization reagent, e.g., a chemical that binds the substrate efficiently and also couples efficiently to monomeric nucleic acid molecules. In some cases, such molecules have a hydroxyl group which is available for interacting with a nucleoside in a coupling reaction.
  • the amino silane is selected from the group consisting of 11-acetoxyundecyltriethoxysilane, n-decyltriethoxysilane, (3-aminopropyl)trimethoxysilane, (3-aminopropyl)triethoxysilane, glycidyloxypropyl/trimethoxysilane and N-(3-triethoxysilylpropyl)-4-hydroxybutyramide.
  • the high surface energy region includes a passive functionalization reagent, e.g., a chemical that binds the substrate efficiently but does not couple efficiently to monomeric nucleic acid molecules.
  • substrates comprising a plurality of clusters, wherein each cluster comprises a plurality of loci that support the attachment and synthesis of oligonucleic acids.
  • a locus is on a three-dimensional surface, e.g., a well, microchannel, channel, or post.
  • a surface of a locus comprises a material that is actively functionalized to attach to at least one nucleotide for oligonucleic acid synthesis, or preferably, a population of identical nucleotides for synthesis of a population of oligonucleic acids.
  • oligonucleic acid refers to a population of oligonucleic acids encoding for the same nucleic acid sequence.
  • a surface of a substrate is inclusive of one or a plurality of surfaces of a substrate.
  • adhesion promoter facilitates structured patterning of loci on a surface of a substrate.
  • exemplary surfaces which can benefit from adhesion promotion include, without limitation, glass, silicon, silicon dioxide and silicon nitride.
  • the adhesion promoter is a chemical with a high surface energy.
  • a second chemical layer is deposited on a surface of a substrate.
  • the second chemical layer has a low surface energy. The surface energy of a chemical layer coated on a surface can facilitate localization of droplets on the surface. Depending on the patterning arrangement selected, the proximity of loci and/or area of fluid contact at the loci can be altered.
  • a substrate surface, or resolved loci, onto which nucleic acids or other moieties are deposited, e.g., for oligonucleic acid synthesis, are smooth or substantially planar or have raised or lowered features.
  • a substrate surface is modified with one or more different layers of compounds.
  • modification layers of interest include, without limitation, inorganic and organic layers such as metals, metal oxides, polymers, small organic molecules and the like.
  • Non-limiting polymeric layers include peptides, proteins, nucleic acids or mimetics thereof (e.g., peptide nucleic acids and the like), polysaccharides, phospholipids, polyurethanes, polyesters, polycarbonates, polyureas, polyamides, polyetheyleneamines, polyarylene sulfides, polysiloxanes, polyimides, polyacetates, and any other suitable compounds described herein or otherwise known in the art.
  • polymers are heteropolymeric.
  • polymers are homopolymeric.
  • polymers comprise functional moieties or are conjugated.
  • resolved loci of a substrate are functionalized with one or more moieties that increase and/or decrease surface energy.
  • a moiety is chemically inert.
  • a moiety is configured to support a desired chemical reaction, for example, one or more processes in an oligonucleic acid synthesis reaction.
  • the surface energy, or hydrophobicity, of a surface is a factor for determining the affinity of a nucleotide to attach onto the surface.
  • a method for substrate functionalization comprises: (a) providing a substrate (e.g., a plate); and (b) silanizing the loci (e.g., microchannels) with a suitable silanizing agent described herein or otherwise known in the art, for example, an organofunctional alkoxysilane molecule.
  • the organofunctional alkoxysilane molecule comprises dimethylchloro-octodecyl-silane, methyldichloro-octodecyl-silane, trichloro-octodecyl-silane, trimethyl-octodecyl-silane, triethyl-octodecyl-silane, or any combination thereof.
  • a substrate surface comprises functionalized with polyethylene/polypropylene (functionalized by gamma irradiation or chromic acid oxidation, and reduction to hydroxyalkyl surface), highly crosslinked polystyrene-divinylbenzene (derivatized by chloromethylation, and aminated to benzylamine functional surface), nylon (the terminal aminohexyl groups are directly reactive), or etched with reduced polytetrafluoroethylene.
  • polyethylene/polypropylene functionalized by gamma irradiation or chromic acid oxidation, and reduction to hydroxyalkyl surface
  • highly crosslinked polystyrene-divinylbenzene derivatized by chloromethylation, and aminated to benzylamine functional surface
  • nylon the terminal aminohexyl groups are directly reactive
  • a substrate surface is functionalized by contact with a derivatizing composition that contains a mixture of silanes, under reaction conditions effective to couple the silanes to the substrate surface, typically via reactive hydrophilic moieties present on the substrate surface.
  • Silanization generally can be used to cover a surface through self-assembly with organofunctional alkoxysilane molecules.
  • a variety of siloxane functionalizing reagents can further be used as currently known in the art, e.g., for lowering or increasing surface energy.
  • the organofunctional alkoxysilanes are classified according to their organic functions.
  • Non-limiting examples of siloxane functionalizing reagents include hydroxyalkyl siloxanes (silylate surface, functionalizing with diborane and oxidizing the alcohol by hydrogen peroxide), diol (dihydroxyalkyl) siloxanes (silylate surface, and hydrolyzing to diol), aminoalkyl siloxanes (amines require no intermediate functionalizing step), glycidoxysilanes (3-glycidoxypropyl-dimethyl-ethoxysilane, glycidoxy-trimethoxysilane), mercaptosilanes (3-mercaptopropyl-trimethoxysilane, 3-4 epoxycyclohexyl-ethyltrimethoxysilane or 3-mercaptopropyl-methyl-dimethoxysilane), bicyclohepthenyl-trichlorosilane, butyl-aldehydr-trimethoxysilane, or dimeric secondary aminoalkyl siloxanes.
  • the hydroxyalkyl siloxanes can include allyl trichlorochlorosilane turning into 3-hydroxypropyl, or 7-oct-1-enyl trichlorochlorosilane turning into 8-hydroxyoctyl.
  • the diol (dihydroxyalkyl) siloxanes include glycidyl trimethoxysilane-derived (2,3-dihydroxypropyloxy)propyl (GOPS).
  • the aminoalkyl siloxanes include 3-aminopropyl trimethoxysilane turning into 3-aminopropyl (3-aminopropyl-triethoxysilane, 3-aminopropyl-diethoxy-methylsilane, 3-aminopropyl-dimethyl-ethoxysilane, or 3-aminopropyl-trimethoxysilane).
  • the dimeric secondary aminoalkyl siloxanes can be bis (3-trimethoxysilylpropyl) amine turning into bis(silyloxylpropyl)amine.
  • the functionalizing agent comprises 11-acetoxyundecyltriethoxysilane, n-decyltriethoxysilane, (3-aminopropyl)trimethoxysilane, (3-aminopropyl)triethoxysilane, glycidyloxypropyl/trimethoxysilane and N-(3-triethoxysilylpropyl)-4-hydroxybutyramide.
  • a substrate surface is contacting with a mixture of functionalization groups, e.g., aminosilanes, which can be in any different ratio.
  • a mixture comprises at least 2, 3, 4, 5 or more different types of functionalization agents.
  • the mixture comprises 1, 2, 3 or more silanes.
  • the ratio of the at least two types of surface functionalization agents in a mixture is about 1:1, 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, 1:10, 2:3, 2:5, 2:7, 2:9, 2:11, 2:13, 2:15, 2:17, 2:19, 3:5, 3:7, 3:8, 3:10, 3:11, 3:13, 3:14, 3:16, 3:17, 3:19, 4:5, 4:7, 4:9, 4:11, 4:13, 4:15, 4:17, 4:19, 5:6, 5:8, 5:9, 5:11, 5:12, 5:13, 5:14, 5:16, 5:17, 5:18, 5:19, 6:7, 6:11, 6:13, 6:17, 6:19, 7:8, 7:9, 7:10, 7:11, 7:12, 7:13, 7:15, 7:16, 7:18, 7:19, 8:9, 8:11, 8:13, 8:15, 8:17, 8
  • an active functionalization agent comprises 11-acetoxyundecyltriethoxysilane. In some cases, an active functionalization agent comprises n-decyltriethoxysilane. In some cases, the active functionalization areas comprise one or more different species of silanes, for example, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more silanes. In some cases, one of the one or more silanes is present in the functionalization composition in an amount greater than another silane.
  • a mixed silane solution having two silanes comprises a 99:1, 98:2, 97:3, 96:4, 95:5, 94:6, 93:7, 92:8, 91:9, 90:10, 89:11, 88:12, 87:13, 86:14, 85:15, 84:16, 83:17, 82:18, 81:19, 80:20, 75:25, 70:30, 65:35, 60:40, 55:45 ratio of one silane to another silane.
  • an active functionalization agent comprises 11-acetoxyundecyltriethoxysilane and n-decyltriethoxysilane (e.g., in a ratio from about 20:80 to about 1:99, or about 10:90 to about 2:98, or preferably about 5:95).
  • an active functionalization agent comprises glycidyloxypropyltriethoxysilane (GOPS).
  • GOPS glycidyloxypropyltriethoxysilane
  • the silane is a fluorosilane.
  • the silane is a hydrocarbon silane.
  • the silane is 3-iodo-propyltrimethoxysilane.
  • the silane is octylchlorosilane.
  • an active functionalization agent comprises N-(3-triethosysilylpropyl)-4-hydroxybutyramide.
  • the passive functionalization agent comprises a silane.
  • the passive functionalization agent comprises a mixture of silanes.
  • the passive functionalization agent comprises perfluorooctyltrichlorosilane.
  • desired surface tensions, wettabilities, water contact angles, and/or contact angles for other suitable solvents are achieved by providing a substrate surface with a suitable ratio of functionalization agents.
  • the agents in a mixture are chosen from suitable reactive and inert moieties, thus diluting the surface density of reactive groups to a desired level for downstream reactions.
  • the density of the fraction of a surface functional group that reacts to form a growing oligonucleotide in an oligonucleotide synthesis reaction is about 0.005, 0.01, 0.05, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1.0, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2.0, 2.1, 2.2, 2.3, 2.4, 2.5, 3.0, 3.5, 4.0, 4.5, 5.0, 7.0, 10.0, 15.0, 20.0, 50.0, 75.0, 100.0 ⁇ Mol/m 2 .
  • a surface of a substrate is prepared to have a low surface energy.
  • a region of a surface of a substrate described herein is functionalized to enable covalent binding of molecular moieties that can lower the surface energy so that wettability can be reduced.
  • a region of surface of a substrate described herein is prepared to have a high surface energy and increased wettability.
  • a surface is modified to have a higher surface energy, or become more hydrophilic with a coating where the coating includes molecules having reactive hydrophilic moieties.
  • a deposited reagent liquid e.g., a reagent deposited during an oligonucleic acid synthesis method
  • a droplet of reagent is deposited over a predetermined area of a surface with high surface energy.
  • the liquid droplet can spread over and fill a small surface area having a higher surface energy as compared to a nearby surface.
  • a substrate surface is modified to comprise reactive hydrophilic moieties such as hydroxyl groups, carboxyl groups, thiol groups, and/or substituted or unsubstituted amino groups.
  • Suitable materials include, but are not limited to, supports that can be used for solid phase chemical synthesis, e.g., cross-linked polymeric materials (e.g., divinylbenzene styrene-based polymers), agarose (e.g., Sepharose®), dextran (e.g., Sephadex®), cellulosic polymers, polyacrylamides, silica, glass (particularly controlled pore glass, or “CPG”), ceramics, and the like.
  • the supports may be obtained commercially and used as is, or they may be treated or coated prior to functionalization.
  • a substrate e.g., a silicon plate
  • a substrate e.g., a silicon plate
  • a first layer of a material that modifies the surface to allow for adhesion of a photoresist.
  • Non-limiting examples of materials for surface modification include 11-acetoxyundecyltriethoxysilane, n-decyltriethoxysilane, (3-aminopropyl)trimethoxysilane, (3-aminopropyl)triethoxysilane, glycidyloxypropyl/trimethoxysilane and N-(3-triethoxysilylpropyl)-4-hydroxybutyramide, and mixtures thereof, and/or other suitable materials described elsewhere herein or known in the art.
  • the modified substrate is treated with resist, exposed, developed, and residual resist is removed by plasma cleaning, exemplary details of which are described previously herein.
  • the substrate is passively functionalized, for example, with fluorosilane, to generate hydrophobic regions.
  • the substrate is stripped to remove remaining resist.
  • the first modified layer of the substrate is activated to change one or more functional groups of the surface modification material to a hydroxyl group.
  • the hydroxyl surface of the substrate is treated with a second layer of surface modification material to dilute surface functional groups and increase coupling efficiency for nucleotide attachment.
  • the second layer is activated to change one or more functional groups to a hydroxyl group to support oligonucleic acid synthesis.
  • steps comprising the addition of a surface modification material followed by subsequent activation are repeated for one or more additional cycles (e.g., 1, 2, 3, 4, 5 or more additional cycles) to provide optimal spacing between oligonucleic acids during synthesis.
  • second and subsequent layers have purely organic chemistries of the general form A-R1-B and C—R2-D where A reacts with the terminal OH group. The system is then purged and C—R2-D reacts with the B group. R1 and R2 chemistries can be repeated to yield the desired film thickness in a molecular layer deposition process.
  • the terminal groups B and D can be hydroxyl or could be converted to hydroxyl in the final step of the deposition.
  • the substrate may be two-dimensional (e.g., substantially planar) or three-dimension (e.g., comprise wells and/or channels).
  • an actively functionalized surface comprises a specific concentration of hydroxyl groups to achieve a pre-determined surface density for oligonucleic acid synthesis.
  • active functionalization is achieved by a wet process using a solution comprising an active functionalization agent.
  • the active functionalization agent comprises a silane or mixed silanes.
  • a surface to be actively functionalized is treated with a solution comprising an active functionalization agent (e.g., 1% solution of N-(3-triethoxysilylpropyl-4hydroxybutyramide in ethanol and acetic acid) and the substrate incubated at a high temperature (e.g., 150° C. for 14 hours).
  • an active functionalization agent e.g., 1% solution of N-(3-triethoxysilylpropyl-4hydroxybutyramide in ethanol and acetic acid
  • a chemical vapor deposition process is employed wherein an active functionalization agent is delivered to the surface in a gaseous state.
  • an active functionalization agent is delivered by CVD with a controlled deposition pressure (e.g., 200 mTor) and temperature (e.g., 150° C.).
  • a CVD process allows for in-situ plasma cleaning and is well suited for producing highly ordered self-assembled monolayers (SAMs).
  • the surface energy, or hydrophobicity of a surface can be evaluated or measured by measuring a water contact angle.
  • Water contact angle is the angle between the drop surface and a solid surface where a water droplet meets the solid surface.
  • a surface with a water contact angle of smaller than 90°, the solid surface can be considered hydrophilic or polar.
  • a surface with a water contact angle of greater than 90° the solid surface can be considered hydrophobic or apolar.
  • the surface is selected to be inert to the conditions of ordinary oligonucleotide synthesis; e.g. the solid surface may be devoid of free hydroxyl, amino, or carboxyl groups to the bulk solvent interface during monomer addition, depending on the selected chemistry.
  • the surface may comprise reactive moieties prior to the start of a first cycle, or first few cycles of an oligonucleotide synthesis process, wherein the reactive moieties can be quickly depleted to unmeasurable densities after one, two, three, four, five, or more cycles of the oligonucleotide synthesis reaction.
  • the surface can further be optimized for well or pore wetting, e.g., by common organic solvents such as acetonitrile and the glycol ethers or aqueous solvents, relative to surrounding surfaces.
  • the wetting phenomenon is understood to be a measure of the surface tension or attractive forces between molecules at a solid-liquid interface, and is expressed in dynes/cm 2 .
  • fluorocarbons have very low surface tension, which is typically attributed to the unique polarity (electronegativity) of the carbon-fluorine bond.
  • surface tension of a layer can be primarily determined by the percent of fluorine in the terminus of the alkyl chains.
  • a single terminal trifluoromethyl group can render a surface nearly as lipophobic as a perfluoroalkyl layer.
  • the density of reactive sites can be lower than Langmuir-Blodgett and group density.
  • surface tension of a methyltrimethoxysilane surface can be about 22.5 mN/m and aminopropyltriethoxysilane surface can be about 35 mN/m.
  • hydrophilic behavior of surfaces is generally considered to occur when critical surface tensions are greater than 45 mN/m. As the critical surface tension increases, the expected decrease in contact angle is accompanied with stronger adsorptive behavior. Hydrophobic behavior of surfaces is generally considered to occur when critical surface tensions are less than 35 mN/m.
  • the decrease in critical surface tension is associated with oleophilic behavior, i.e. the wetting of the surfaces by hydrocarbon oils.
  • the critical surface tensions decrease below 20 mN/m
  • the surfaces resist wetting by hydrocarbon oils and are considered both oleophobic as well as hydrophobic.
  • silane surface modification can be used to generate a broad range of critical surface tensions.
  • the methods and compositions of the invention may use surface coatings, e.g. those involving silanes, to achieve surface tensions of less than 5, 6, 7, 8, 9, 10, 12, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 110, 115, 120 mN/m, or higher.
  • the methods and compositions of the invention may use surface coatings, e.g. those involving silanes, to achieve surface tensions of more than 115, 110, 100, 90, 80, 70, 60, 50, 45, 40, 35, 30, 25, 20, 15, 12, 10, 9, 8, 7, 6 mN/m or less.
  • surface coatings e.g. those involving silanes
  • the water contact angle and the surface tension of non-limiting examples of surface coatings, e.g., those involving silanes are described in Table 1 and Table 2 of Arkles et al. (Silanes and Other Coupling Agents, Vol. 5v: The Role of Polarity in the Structure of Silanes Employed in Surface Modification. 2009), which is incorporated herein by reference in its entirety. The tables are replicated below.
  • the surface of the substrate or a portion of the surface of the substrate can be functionalized or modified to be more hydrophilic or hydrophobic as compared to the surface or the portion of the surface prior to the functionalization or modification.
  • one or more surfaces can be modified to have a difference in water contact angle of greater than 90°, 85°, 80°, 75°, 70°, 65°, 60°, 55°, 50°, 45°, 40°, 35°, 30°, 25°, 20°, 15° or 10° as measured on one or more uncurved, smooth or planar equivalent surfaces.
  • the surface of the microstructures, channels, resolved loci, resolved reactor caps or other parts of the substrate may be modified to have a differential hydrophobicity corresponding to a difference in water contact angle that is greater than 90°, 85°, 80°, 75°, 70°, 65°, 60°, 55°, 50°, 45°, 40°, 35°, 30°, 25°, 20°, 15° or 10° as measured on uncurved, smooth or planar equivalent surfaces of such structures.
  • water contact angles mentioned herein correspond to measurements that would be taken on uncurved, smooth or planar equivalents of the surfaces in question.
  • hydrophilic resolved loci can be generated by first applying a protectant, or resist, over each loci within the substrate.
  • the unprotected area can be then coated with a hydrophobic agent to yield an unreactive surface.
  • a hydrophobic coating can be created by chemical vapor deposition of (tridecafluorotetrahydrooctyl)-triethoxysilane onto the exposed oxide surrounding the protected channels or wells.
  • the protectant, or resist can be removed exposing the loci regions of the substrate for further modification and oligonucleotide synthesis.
  • the initial modification of such unprotected regions may resist further modification and retain their surface functionalization, while newly unprotected areas can be subjected to subsequent modification steps.
  • a process for carving features out of a substrate may include providing a substrate having a device layer and a handle layer, wherein the device layer is optionally separated from the handle layer by an electrical insulator layer, e.g., a layer of silicon dioxide—an exemplary substrate is a SOI wafer.
  • an electrical insulator layer e.g., a layer of silicon dioxide—an exemplary substrate is a SOI wafer.
  • the provided substrate is oxidized on both surfaces.
  • Photolithography may be applied to the device side of the substrate to create a mask of photoresist.
  • a deep reactive-ion etching (DRIE) step may be used to etch vertical side-walls (e.g., until an insulator layer in a substrate comprising an insulator layer) at locations devoid of the photoresist.
  • DRIE deep reactive-ion etching
  • the photoresist may be stripped.
  • photolithography, DRIE and photoresist strip steps are repeated on the substrate handle side.
  • the substrate comprises an insulator layer such as silicon dioxide
  • buried oxide (BOX) may be removed using an etching process.
  • Thermal oxidation can then be applied to remove contaminating polymers that may have been deposited on the side walls during the method.
  • the thermal oxidation may be stripped using a wet etching process. In some instances, this method is used to generate a substrate having the features of a substrate exemplified in FIGS. 4 A- 4 B .
  • the process for manufacturing a substrate comprises a front-end process method comprising providing a starting material substrate, oxidizing both the device and handle sides; performing photolithography, DRIE and stripping of photoresist on the handle side; performing photolithography, DRIE and stripping of photoresist on the device side; removal of the oxide layer (e.g., BOX); and oxide growth (e.g., oxide is coated on one or more surfaces of the substrate to create a silicon substrate having a plurality of features).
  • oxide layer e.g., BOX
  • oxide growth e.g., oxide is coated on one or more surfaces of the substrate to create a silicon substrate having a plurality of features.
  • the substrate starting material comprises silicon.
  • the substrate is oxidized on one or more surfaces.
  • photolithography is applied to the front-side, back-side or both the front and back sides of the substrate, for example, to the back-side, to create a mask of photoresist.
  • the substrate is etched at locations devoid of photoresist, in many cases, beyond the oxidized layer, to create wells.
  • the back-side is etched.
  • the photoresist is stripped.
  • a second side of the substrate is subjected to photolithography.
  • the back-side was first subjected to photolithography followed by photolithography of the front-side of the substrate.
  • a deep reactive-ion etching (DRIE) is used to etch vertical side walls to a prescribed depth, for example, about 450 um.
  • DRIE is used on the front-side, back-side or both the front and back sides during photolithography.
  • only one side of a substrate is etched to create three-dimensional features.
  • two sides, e.g., device and handle sides, of a substrate is etched to create three-dimensional features.
  • a SOI substrate silicon on insulator silicon wafer
  • the handle layer is etched down to the buried oxide, wherein the buried oxide can serve as an etch stop.
  • the photoresist is stripped to generate a desired pattern.
  • the front-end is resist stripped to generate three dimensional features.
  • contaminates of the process e.g., fluoropolymers
  • the substrate processed may comprise a plurality of wells, where the distance from the center of one main channel to the center of another main channel is about 1.69 mm and the total height of the main channel/microchannel is about 450 um.
  • a substrate described herein may comprise fiducial marks to facilitate alignment with other components of a system.
  • substrates have one or more fiducial marks, e.g. 2, 3, 4, 5, 6, 7, 8, 9, 10, or more fiducial marks.
  • a fiducial mark may be located near the origin, where the fiducial mark is closer to the origin than any one cluster.
  • a fiducial mark is located near an edge of the substrate portion.
  • the fiducial mark may be located from about 0.1 mm to about 10 mm from the edge of the substrate portion, e.g., about 0.5 mm from the edge.
  • the fiducial mark may be located close to or distant to a cluster.
  • a fiducial is located from about 1 mm to about 10 mm form a cluster, e.g., 1.69 mm.
  • a distance from the center of a fiducial mark and a nearest corner of a substrate in one dimension is from about 0.5 mm to about 10 mm, e.g., about 1 mm.
  • a length of a fiducial mark in one dimension is from about 0.5 mm to about 5 mm, e.g., about 1 mm.
  • the width of a fiducial mark is from about 0.01 mm to about 2 mm, e.g., 0.05 mm.
  • the substrates described herein, in some instances, comprise one or more regions for annotation.
  • a substrate may have a label or serial number which is located a distance (e.g., 4 mm) from the edge of the substrate with a length (e.g., 9 mm) and width (e.g., 1.5 mm).
  • Structures having modified surfaces described herein may be used for de novo synthesis processes.
  • An exemplary workflow for one such process is divided generally into phases: (1) de novo synthesis of a single stranded oligonucleic acid library, (2) joining oligonucleic acids to form larger fragments, (3) error correction, (4) quality control, and (5) shipment, FIG. 12 .
  • an intended nucleic acid sequence or group of nucleic acid sequences is preselected. For example, a group of genes is preselected for generation.
  • a predetermined library of oligonucleic acids is designed for de novo synthesis.
  • Various suitable methods are known for generating high density oligonucleic acid arrays.
  • a surface layer 1201 is provided.
  • chemistry of the surface is altered in order to improve the oligonucleic acid synthesis process. Areas of low surface energy are generated to repel liquid while areas of high surface energy are generated to attract liquids.
  • the surface itself may be in the form of a planar surface or contain variations in shape, such as protrusions, microwells, or microchannels which increase surface area.
  • high surface energy molecules selected serve a dual function of supporting DNA chemistry, as described in International Patent Application Publication WO/2015/021280, which is herein incorporated by reference in its entirety.
  • oligonucleic acid arrays In situ preparation of oligonucleic acid arrays is generated on a solid support and utilizes single nucleotide extension process to extend multiple oligomers in parallel.
  • a device such as an oligonucleic acid synthesizer, is designed to release reagents in a step wise fashion such that multiple oligonucleic acids extend, in parallel, one residue at a time to generate oligomers with a predetermined nucleic acid sequence 1202 .
  • oligonucleic acids are cleaved from the surface at this stage. Cleavage may include gas cleavage, e.g., with ammonia or methylamine.
  • the generated oligonucleic acid libraries are placed in a reaction chamber.
  • the reaction chamber also referred to as “nanoreactor” is a silicon coated main channel, containing PCR reagents and lowered onto the oligonucleic acid library 1203 .
  • a reagent is added to release the oligonucleic acids from the surface.
  • the oligonucleic acids are released subsequent to sealing of the nanoreactor 1205 . Once released, fragments of single stranded oligonucleic acids hybridize in order to span an entire long range sequence of DNA. Partial hybridization 1205 is possible because each synthesized oligonucleic acid is designed to have a small portion overlapping with at least one other oligonucleic acid in the pool.
  • a PCA reaction is commenced.
  • the oligonucleic acids anneal to complementary fragments and gaps are filled in by a polymerase.
  • Each cycle increases the length of various fragments randomly depending on which oligonucleic acids find each other. Complementarity amongst the fragments allows for forming a complete large span of double stranded DNA 1206 .
  • the nanoreactor is separated from the surface 1207 and positioned for interaction with a polymerase 1208 .
  • the nanoreactor is subject to PCR 1209 and the larger nucleic acids are formed.
  • PCR 1210 the nanochamber is opened 1211 , error correction reagents are added 1212 , the chamber is sealed 1213 and an error correction reaction occurs to remove mismatched base pairs and/or strands with poor complementarity from the double stranded PCR amplification products 1214 .
  • the nanoreactor is opened and separated 1215 . Error corrected product is next subject to additional processing steps, such as PCR and molecular bar coding, and then packaged 1222 for shipment 1223 .
  • quality control measures are taken.
  • quality control steps include for example interaction with a wafer having sequencing primers for amplification of the error corrected product 1216 , sealing the wafer to a chamber containing error corrected amplification product 1217 , and performing an additional round of amplification 1218 .
  • the nanoreactor is opened 1219 and the products are pooled 1220 and sequenced 1221 . After an acceptable quality control determination is made, the packaged product 1222 is approved for shipment 1223 .
  • Oligonucleic acids may be synthesized on a substrate described herein using a system comprising an oligonucleic acid synthesizer that deposits reagents necessary for synthesis.
  • Reagents for oligonucleic acid synthesis include, for example, reagents for oligonucleic acid extension and wash buffers.
  • the oligonucleic acid synthesizer deposits coupling reagents, capping reagents, oxidizers, de-blocking agents, acetonitrile and gases such as nitrogen gas.
  • the oligonucleic acid synthesizer optionally deposits reagents for the preparation and/or maintenance of substrate integrity.
  • An oligonucleic acid synthesizer may comprise material deposition devices that can move in the X-Y direction to align with the location of the substrate.
  • the oligonucleic acid synthesizer can also move in the Z direction to seal with the substrate, forming a resolved reactor.
  • a substrate having a plurality of clusters is configured to seal with a capping element having a plurality of caps, wherein when the substrate and capping element are sealed, each cluster is separate from another cluster to form separate resolved reactors for each cluster.
  • the capping element is not present in the system or is present and stationary.
  • a resolved reactor is configured to allow for the transfer of fluid, including oligonucleic acids and/or reagents, from the substrate to the capping element and/or vice versa.
  • Fluid may pass through either or both the substrate and the capping element and includes, without limitation, coupling reagents, capping reagents, oxidizers, de-blocking agents, acetonitrile and nitrogen gas.
  • the oligonucleic acid synthesizer of an oligonucleic acid synthesis system may comprise a plurality of material deposition devices, for example from about 1 to about 50 material deposition devices. Each material deposition device, in various instances, deposits a reagent component that is different from another material deposition device.
  • each material deposition device has a plurality of nozzles, where each nozzle is optionally configured to correspond to a cluster on a substrate. For example, for a substrate having 256 clusters, a material deposition device has 256 nozzles and 100 ⁇ m fly height. In some cases, each nozzle deposits a reagent component that is different from another nozzle.
  • the substrates described herein comprise actively functionalized surfaces configured to support the attachment and synthesis of oligonucleic acids.
  • Synthesized oligonucleic acids include oligonucleic acids comprising modified and/or non-canonical bases and/or modified backbones.
  • a library of oligonucleic acids having pre-selected sequences is synthesized on a substrate.
  • one or more of the oligonucleic acids has a different sequence and/or length than another oligonucleic acid in the library.
  • the stoichiometry of each oligonucleic acid synthesized on a substrate is controlled and tunable by varying one or more features of the substrate (e.g., functionalized surface) and/or oligonucleic acid sequence to be synthesized; one or more methods for substrate functionalization and/or oligonucleic acid synthesis; or a combination thereof.
  • controlling the density of a growing oligonucleic acid on a resolved locus of a substrate allows for oligonucleic acids to be synthesized with a low error rate.
  • oligonucleic acid synthesis methods comprise coupling a linker to a surface of a substrate, for example, to an actively functionalized surface of a substrate.
  • a linker separates a synthesized oligonucleic acid from a surface of the substrate.
  • a linker includes a cleavable linker, such as a photocleavable linker.
  • a synthesized oligonucleic acid comprises a cleavable moiety that is introduced during synthesis. In some cases, a synthesized oligonucleic acid does not comprise a linker.
  • the synthesized oligonucleic acid is separated from the linker by one or more cleavable moieties.
  • the synthesized oligonucleic acid comprises a primer and/or adapter sequence that connects to a linker.
  • a linker comprises one or more bases coupled to the surface of a substrate and a cleavable moiety, wherein the cleavable moiety is configured to connect to the synthesized oligonucleic acid.
  • a linker is referred to as a tether or a tether region.
  • a linker comprises about or at least about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50 or more bases located between a surface of a substrate and a synthesized oligonucleic acid.
  • a linker is synthesized and extends 12 to 25 bases from a device surface.
  • a linker comprises a cleavable moiety, wherein the cleavable moiety is a modified or non-canonical base.
  • the cleavable moiety is referred to as a universal moiety.
  • cleavable moieties include, without limitation, thymidine-succinyl hexamide CED phosphoramidite and DMMA.
  • a cleavable moiety is gas cleavable.
  • the linker comprises thymidine-succinyl hexamide CED phosphoramidite or DMMA.
  • the linker comprises a photocleavable primer.
  • a photocleavable linker allows for the synthesized oligonucleic acid to be removed from the substrate without cleaving the protecting groups on the nitrogenous functionalities of each base, for example, by irradiation with light at about 350 nm.
  • Oligonucleic acids synthesized using the methods and/or substrates described herein comprise, in various instances, at least about 50, 60, 70, 75, 80, 90, 100, 120, 150, 200, 300, 400, 500, 600, 700, 800 or more bases.
  • a library of oligonucleic acids is synthesized, wherein a population of distinct oligonucleic acids are assembled to generate a larger nucleic acid comprising at least about 500; 1,000; 2,000; 3,000; 4,000; 5,000; 6,000; 7,000; 8,000; 9,000; 10,000; 11,000; 12,000; 13,000; 14,000; 15,000; 16,000; 17,000; 18,000; 19,000; 20,000; 25,000; 30,000; 40,000; or 50,000 bases.
  • oligonucleic acid synthesis methods described herein are useful for the generation of an oligonucleic acid library comprising at least 500; 1,000; 5,000; 10,000; 20,000; 50,000; 100,000; 200,000; 300,000; 400,000; 500,000; 600,000; 700,000; 800,000; 900,000; 1,000,000; 1,100,000; 1,200,000; 1,300,000; 1,400,000; 1,500,000; 1,600,000; 1,700,000; 1,800,000; 1,900,000; 2,000,000; 2,200,000; 2,400,000; 2,600,000; 2,800,000; 3,000,000; 3,500,000; 4,00,000; or 5,000,000 distinct oligonucleic acids.
  • At least about 1 ⁇ mol, 10 pmol, 20 pmol, 30 pmol, 40 pmol, 50 pmol, 60 pmol, 70 pmol, 80 pmol, 90 pmol, 100 pmol, 150 pmol, 200 pmol, 300 pmol, 400 pmol, 500 pmol, 600 pmol, 700 pmol, 800 pmol, 900 pmol, 1 nmol, 5 nmol, 10 nmol, 100 nmol or more of an oligonucleic acid is synthesized within a locus.
  • Methods for oligonucleic acid synthesis on a surface allow for synthesis at a fast rate.
  • at least 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 35, 40, 45, 50, 55, 60, 70, 80, 90, 100, 125, 150, 175, 200 nucleotides per hour, or more are synthesized.
  • Nucleotides include adenine, guanine, thymine, cytosine, uridine building blocks, or analogs/modified versions thereof.
  • libraries of oligonucleic acids are synthesized in parallel on substrate.
  • a substrate comprising about or at least about 100; 1,000; 10,000; 100,000; 1,000,000; 2,000,000; 3,000,000; 4,000,000; or 5,000,000 resolved loci is able to support the synthesis of at least the same number of distinct oligonucleic acids, wherein oligonucleic acid encoding a distinct sequence is synthesized on a resolved locus.
  • a library of oligonucleic acids are synthesized on a substrate with low error rates described herein in less than about three months, two months, one month, three weeks, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2 days, 24 hours, 18 hours, 12 hours or less.
  • nucleic acids assembled from an oligonucleic acid library synthesized with low error rate using the substrates and methods described herein are prepared in less than about three months, two months, one month, three weeks, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2 days, 24 hours, 18 hours, 12 hours or less.
  • up to about 800,000 distinct oligonucleic acids having sizes up to about 130 base pairs in length each are synthesized with an error rate below 1:1000, 1:2000, 1:3000 or less on a substrate described herein and using a method described herein in a span of less than about 24 hours.
  • oligonucleic acid error rate is dependent on the efficiency of one or more chemical steps of oligonucleic acid synthesis.
  • oligonucleic acid synthesis comprises a phosphoramidite method, wherein a base of a growing oligonucleic acid chain is coupled to phosphoramidite.
  • coupling efficiency of the base is related to error rate. For example, higher coupling efficiency correlates to lower error rates.
  • an oligonucleic acid synthesis method comprises a double coupling process, wherein a base of a growing oligonucleic acid chain is coupled with a phosphoramidite, the oligonucleic acid is washed and dried, and then treated a second time with a phosphoramidite.
  • efficiency of deblocking in a phosphoramidite oligonucleic acid synthesis method contributes to error rate.
  • the substrates and/or synthesis methods described herein allow for removal of 5′-hydroxyl protecting groups at efficiencies greater than 98%, 98.5%, 99%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9%, 99.95%, 99.96%, 99.97%, 99.98%, or 99.99%.
  • error rate is reduced by minimization of depurination side reactions.
  • Oligonucleic acids synthesized using the methods and/or substrates described herein encode for, in various instances, at least about 50, 60, 70, 75, 80, 90, 100, 120, 150, 200, 240, 300, 400, 500, 600, 700, 800, 900, 1,000, 6000, 6144, 10,000, or more genes.
  • a library of oligonucleic acids encode for at least 200 genes.
  • a library of oligonucleic acids encode for genes at least 500 bases, 1 kb, 2 kb, 3 kb, 4 kb, 5 kb or more in length.
  • oligonucleic acid synthesis comprises coupling a base with phosphoramidite. In some instances, oligonucleic acid synthesis comprises coupling a base by deposition of phosphoramidite under coupling conditions, wherein the same base is optionally deposited with phosphoramidite more than once, i.e., double coupling. In some instances, oligonucleic acid synthesis comprises capping of unreacted sites. In some cases, capping is optional. In some instances, oligonucleic acid synthesis comprises oxidation. In some instances, oligonucleic acid synthesis comprises deblocking or detritylation.
  • oligonucleic acid synthesis comprises sulfurization. In some cases, oligonucleic acid synthesis comprises either oxidation or sulfurization. In some instances, between one or each step during an oligonucleic acid synthesis reaction, the substrate is washed, for example, using tetrazole or acetonitrile. Time frames for any one step in a phosphoramidite synthesis method include less than about 2 min, 1 min, 50 sec, 40 sec, 30 sec, 20 sec and 10 sec.
  • Oligonucleic acid synthesis using a phosphoramidite method comprises the subsequent addition of a phosphoramidite building block (e.g., nucleoside phosphoramidite) to a growing oligonucleic acid chain for the formation of a phosphite triester linkage.
  • Phosphoramidite oligonucleic acid synthesis proceeds in the 3′ to 5′ direction.
  • Phosphoramidite oligonucleic acid synthesis allows for the controlled addition of one nucleotide to a growing nucleic acid chain per synthesis cycle. In some instances, each synthesis cycle comprises a coupling step.
  • Phosphoramidite coupling involves the formation of a phosphite triester linkage between an activated nucleoside phosphoramidite and a nucleoside bound to the substrate, for example, via a linker.
  • the nucleoside phosphoramidite is provided to the substrate activated.
  • the nucleoside phosphoramidite is provided to the substrate with an activator.
  • nucleoside phosphoramidites are provided to the substrate in a 1.5, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100-fold excess or more over the substrate-bound nucleosides.
  • nucleoside phosphoramidite is performed in an anhydrous environment, for example, in anhydrous acetonitrile.
  • the substrate is optionally washed.
  • the coupling step is repeated one or more additional times, optionally with a wash step between nucleoside phosphoramidite additions to the substrate.
  • an oligonucleic acid synthesis method used herein comprises 1, 2, 3 or more sequential coupling steps.
  • the nucleoside bound to the substrate is de-protected by removal of a protecting group, where the protecting group functions to prevent polymerization.
  • a common protecting group is 4,4′-dimethoxytrityl (DMT).
  • phosphoramidite oligonucleic acid synthesis methods optionally comprise a capping step.
  • a capping step the growing oligonucleic acid is treated with a capping agent.
  • a capping step is useful to block unreacted substrate-bound 5 ′—OH groups after coupling from further chain elongation, preventing the formation of oligonucleic acids with internal base deletions.
  • phosphoramidites activated with 1H-tetrazole may react, to a small extent, with the O6 position of guanosine. Without being bound by theory, upon oxidation with I 2 /water, this side product, possibly via O6-N7 migration, may undergo depurination.
  • the apurinic sites may end up being cleaved in the course of the final deprotection of the oligonucleotide thus reducing the yield of the full-length product.
  • the O6 modifications may be removed by treatment with the capping reagent prior to oxidation with I 2 /water.
  • inclusion of a capping step during oligonucleic acid synthesis decreases the error rate as compared to synthesis without capping.
  • the capping step comprises treating the substrate-bound oligonucleic acid with a mixture of acetic anhydride and 1-methylimidazole. Following a capping step, the substrate is optionally washed.
  • the substrate bound growing nucleic acid is oxidized.
  • the oxidation step comprises the phosphite triester is oxidized into a tetracoordinated phosphate triester, a protected precursor of the naturally occurring phosphate diester internucleoside linkage.
  • oxidation of the growing oligonucleic acid is achieved by treatment with iodine and water, optionally in the presence of a weak base (e.g., pyridine, lutidine, collidine). Oxidation may be carried out under anhydrous conditions.
  • Oxidation may be carried out using, for example, tert-Butyl hydroperoxide or (1S)-(+)-(10-camphorsulfonyl)-oxaziridine (CSO).
  • CSO tert-Butyl hydroperoxide
  • a capping step is performed following oxidation.
  • a second capping step allows for substrate drying, as residual water from oxidation that may persist can inhibit subsequent coupling.
  • the substrate and growing oligonucleic acid is optionally washed.
  • the step of oxidation is substituted with a sulfurization step to obtain oligonucleotide phosphorothioates, wherein any capping steps can be performed after the sulfurization.
  • reagents are capable of the efficient sulfur transfer, including but not limited to, 3-(dimethylaminomethylidene)amino)-3H-1,2,4-dithiazole-3-thione, DDTT, 3H-1,2-benzodithiol-3-one 1,1-dioxide, also known as Beaucage reagent, and N,N,N′N′-tetraethylthiuram disulfide (TETD).
  • DDTT 3-(dimethylaminomethylidene)amino)-3H-1,2,4-dithiazole-3-thione
  • DDTT 3H-1,2-benzodithiol-3-one 1,1-dioxide
  • Beaucage reagent also known as Beaucage reagent
  • TETD N,N,N′N′-tetraethylthiuram disulfide
  • the protected 5′ end of the substrate bound growing oligonucleic acid must be removed so that the primary hydroxyl group can react with a next nucleoside phosphoramidite.
  • the protecting group is DMT and deblocking occurs with trichloroacetic acid in dichloromethane. Conducting detritylation for an extended time or with stronger than recommended solutions of acids may lead to increased depurination of solid support-bound oligonucleotide and thus reduces the yield of the desired full-length product.
  • Methods and compositions of the invention described herein provide for controlled deblocking conditions limiting undesired depurination reactions.
  • the substrate bound oligonucleic acid is washed after deblocking. In some cases, efficient washing after deblocking contributes to synthesized oligonucleic acids having a low error rate.
  • Methods for the synthesis of oligonucleic acids typically involve an iterating sequence of the following steps: application of a protected monomer to an actively functionalized surface (e.g., locus) to link with either the activated surface, a linker or with a previously deprotected monomer; deprotection of the applied monomer so that it can react with a subsequently applied protected monomer; and application of another protected monomer for linking.
  • One or more intermediate steps include oxidation or sulfurization.
  • one or more wash steps precede or follow one or all of the steps.
  • Methods for phosphoramidite based oligonucleic acid synthesis comprise a series of chemical steps.
  • one or more steps of a synthesis method involve reagent cycling, where one or more steps of the method comprise application to the substrate of a reagent useful for the step.
  • reagents are cycled by a series of liquid deposition and vacuum drying steps.
  • substrates comprising three-dimensional features such as wells, microwells, channels, microchannels and the like
  • reagents are optionally passed through one or more regions of the substrate via the wells and/or channels.
  • reagents are passed through the substrate during synthesis.
  • reagents are passed horizontally through the substrate.
  • reagents are passed vertically through the substrate. In some instances, reagents are passed over a substrate having curved features to enhance flow. In some cases, reagents are delivered to the substrate through the use of photoresist. In some instances, reagents are delivered to the substrate without moving the substrate. For example, reagents are passed over resolved loci within the substrate by flowing them through the substrate from one surface to an opposite surface of the substrate. In some instances, the substrate is moved, for example, to a flow cell, for reagent application, where it is then optionally repositioned.
  • the substrate is deposited with a nucleoside using an oligonucleic acid synthesizer, moved to a flow cell for treating the substrate to one or more select reagents, and then repositioned back to the oligonucleic acid synthesizer for deposition of a subsequent monomer.
  • Reagent delivery approaches suitable for the synthesis methods of the disclosure include manual and automatic, including use of robotic devices and pulse jets.
  • Reagents include any component of an oligonucleic acid synthesis method, including chemical moieties such as nucleosides, washing solutions, and gases such as nitrogen.
  • one or more reagents applied to the surface of a substrate during oligonucleic acid synthesis comprise a solvent.
  • a solvent comprises propylene carbonate.
  • a solvent comprises 2-methylglutaronitrile and/or 3-methoxypropionitrile.
  • a solvent comprises glutaronitrile.
  • a solvent comprises adiponitrile.
  • the solvent allows for high surface tension for reagent deposition. In some instances, the solvent allows for low surface tension for reagent deposition.
  • the volume of reagents applied to a surface of substrate during oligonucleic acid synthesis is selected on the size, location and/or density of the surface to which the reagent is applied (e.g., an actively functionalized locus).
  • the volume of a drop of reagent applied to a surface during oligonucleic acid synthesis is less than about 0.5 picoliters (pL), 1 pL, 5 pL, 10 pL, 50 pL, 100 pL, 500 pL, 1000 pL, 5000 pL, 10000 pL, 100000 pL, 1000000 pL or 10000000 pL.
  • the reagents are delivered in droplets that have a total volume of about 47 pL or less. In some instances, the reagents are delivered in droplets that have a total volume of about 30 to 50 pL. In some instances, the reagents are delivered in droplets that have a total volume of about 50, 49, 48, 47, 46, 44, 45, 43, 42, 41, 40, 39, 38, 37, 36, 35, 34, 33, 32, 31, 30, 29, 28, 27, 26, or 25 pL. In some instances, the rate at which a drop of reagent is applied is at least about 0.5, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 30, 50 or 100 m/sec.
  • Oligonucleic acid synthesis methods include methods for the application of reagents during one or more steps during synthesis. Controlled application of reagents, such as nucleoside monomers to distinct regions of a substrate is important to achieve low error rates.
  • a reagent is deposited directly into a microchannel, with little or no contamination to an adjacent microchannel.
  • the volume of a reagent to be deposited within a three-dimensional feature such as a well or channel is adjusted to a small enough size to minimize cross-contamination.
  • the reagents are delivered in droplets that have a diameter of less than about 5, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190 or 200 um.
  • a non-limiting method to reduce cross-contamination includes bringing the device that deposits the reagent sufficiently close to the surface such the deposited droplet falls substantially within the selected feature.
  • washing during oligonucleic acid synthesis includes one or all wash steps performed during oligonucleic acid synthesis.
  • a wash step is performed wherein at least or about 60%, 70, 80, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99 or 100% of unincorporated nucleosides or extension reaction reagent are removed from the surface of the substrate.
  • a wash step is performed wherein at least or about 95.0, 95.1, 95.2, 95.3, 95.4, 95.5, 95.6, 95.7, 95.8, 95.9, 96.0, 96.1, 96.2, 96.3, 96.4, 96.5, 96.6, 96.7, 96.8, 96.9, 97.0, 97.1, 97.2, 97.3, 97.4, 97.5, 97.6, 97.7, 97.8, 97.9, 98.0, 98.1, 98.2, 98.3, 98.4, 98.5, 98.6, 98.7, 98.8, 98.9, 99.0, 99.1, 99.2, 99.3, 99.4, 99.5, 99.6, 99.7, 99.8, 99.9, or 100% of unincorporated nucleosides or extension reaction reagent are removed from the surface of the substrate.
  • the configuration of a substrate contributes to wash efficiency and error rate.
  • substrates having channels are washed by passage of a wash solution through the substrate, minimizing fluid contact of the growing oligonucleic acids.
  • the geometry of fluid flow during washing controls interfacial instability.
  • a substrate that is substantial planar, or two-dimensional may have a curved surface to enhance wash efficiency and therefore error rate.
  • optimized wash conditions include those that minimize contact time between the wash reagent and the growing oligonucleic acid.
  • the passage of a wash solution through three-dimensional features allows for the effect washing of all surfaces in a short period of time.
  • a water contact angle for the substrate may be chosen in order to reduce depurination and/or speed of synthesis.
  • lower amount of depurination may be achieved on surfaces of higher surface energy, i.e. lower contact angle.
  • depurination occurs at a rate less than 0.1%, 0.05%, 0.01%, 0.005%, 0.001%, 0.0005%, or 0.0001%.
  • the surface properties of the substrate change during oligonucleic acid synthesis.
  • the substrate at the beginning of synthesis can be relatively hydrophobic and while synthesis proceeds, may become increasingly hydrophilic.
  • Oligonucleic acid features can gain substantial surface energy with increasing oligonucleotide length. Generally, these sites or features consisting of protected oligonucleotide acquire enough surface energy to become spontaneously wet to high surface tension organic solvents commonly used in phosphoramidite synthesis, such as acetonitrile or propylene carbonate, after about 10-20 synthesis cycles.
  • the methods and compositions described allow for varying parameters, such as time, flow rate, temperature, volume, viscosity, and/or reagent concentration, during the synthesis of a growing oligonucleic acid as a function of length to account for the changing surface properties on loci of the surface.
  • parameters such as time, flow rate, temperature, volume, viscosity, and/or reagent concentration
  • Such a variation may be applied by changing parameters in constant or varying increments at repeating cycles of the synthesis.
  • parameters may be changed at only selected cycles of the synthesis and can optionally follow a pattern, such as every other cycle, every third, fourth, fifth, sixth, seventh, eighth, ninth, tenth cycle etc.
  • Oligonucleic acid synthesis methods described herein are suitable for the spatial control of oligonucleic acid synthesis within a small area of a substrate, e.g., a locus.
  • oligonucleic acid methods comprise phosphoramidite chemistry.
  • spatial control of oligonucleic acid synthesis is achieved using an oligonucleic acid synthesizer.
  • spatial control of oligonucleic acid synthesis is achieved using physical masks.
  • spatial control of oligonucleic acid synthesis is achieved by modulation of a 5′ hydroxyl deblocking during phosphoramidite synthesis.
  • spatial control of oligonucleic acid synthesis is achieved by photolithographic deprotection of photolabile monomers. In some instances, spatial control of oligonucleic acid synthesis is achieved by digital activation of photogenerated acids to carry out standard detritylation.
  • the surface of the substrate that provides support for oligonucleic acid synthesis is chemically modified to allow for the synthesized oligonucleic acid chain to be cleaved from the surface.
  • the oligonucleic acid chain is cleaved at the same time as the oligonucleic acid is deprotected. In some cases, the oligonucleic acid chain is cleaved after the oligonucleic acid is deprotected.
  • a trialkoxysilyl amine e.g., (CH 3 CH 2 O) 3 Si—(CH 2 ) 2 —NH 2
  • succinic anhydride e.g., (CH 3 CH 2 O) 3 Si—(CH 2 ) 2 —NH 2
  • oligonucleic acids are synthesized with photolabile protecting groups, where the hydroxyl groups generated on the surface are blocked by photolabile-protecting groups.
  • photolabile protecting groups where the hydroxyl groups generated on the surface are blocked by photolabile-protecting groups.
  • a pattern of free hydroxyl groups on the surface may be generated. These hydroxyl groups can react with photoprotected nucleoside phosphoramidites, according to phosphoramidite chemistry.
  • a second photolithographic mask can be applied and the surface can be exposed to UV light to generate second pattern of hydroxyl groups, followed by coupling with 5′-photoprotected nucleoside phosphoramidite.
  • patterns can be generated and oligomer chains can be extended.
  • the lability of a photocleavable group depends on the wavelength and polarity of a solvent employed and the rate of photocleavage may be affected by the duration of exposure and the intensity of light.
  • This method can leverage a number of factors, e.g., accuracy in alignment of the masks, efficiency of removal of photo-protecting groups, and the yields of the phosphoramidite coupling step. Further, unintended leakage of light into neighboring sites can be minimized.
  • the density of synthesized oligomer per spot can be monitored by adjusting loading of the leader nucleoside on the surface of synthesis.
  • error rate may also be referred to herein as a comparison of the collective sequence encoded by oligonucleic acids generated compared to the aggregate sequence of a predetermined longer nucleic acid, e.g., a gene. Oligonucleic acids are synthesized on a substrate described herein in a process that minimizes the error rate. For example, error rate is less than 1 in 500 bases, 1 in 1000 bases, 1 in 1500 bases, 1 in 2000 bases, 1 in 2500 bases, 1 in 3000, 1 in 5000 bases or less.
  • low error rates are achieved for synthesized oligonucleic acid libraries having at least 20,000; 40,000; 60,000; 80,000; 100,000; 200,000; 300,000; 400,000; 500,000; 600,000; 700,000; 800,000; 1,000,000; or 2,000,000 or more oligonucleic acids.
  • a subset of oligonucleic acids in the library has the same sequence.
  • one or more of the oligonucleic acids in the library comprises a different sequence.
  • Error rates include mismatch error rate, deletion error rate, insertion error rate, indel error rate, and any combination thereof.
  • low overall error rate or low error rates for individual types of errors are achieved.
  • Individual types of error rates include deletions, insertions, or substitutions for an oligonucleic acid library synthesized on the substrate.
  • oligonucleic acids synthesized on the substrate have an average error rate of about 1:500, 1:1000, 1:2000, 1:3000, 1:4000, 1:5000, 1:6000, 1:7000, 1:8000, 1:9000, 1:10000, 1:20000, 1:30000, 1:40000, 1:50000 or less.
  • these error rates are for at least 50%, 60%, 70%, 80%, 90%, 95%, 98%, 99%, 99.5%, or more of the oligonucleic acids synthesized.
  • these error rates are for 100% of the oligonucleic acids synthesized.
  • oligonucleic acids synthesized on the substrate have an average deletion error rate of about 1:500, 1:1000, 1:2000, 1:3000, 1:4000, 1:5000, 1:6000, 1:7000, 1:8000, 1:9000, 1:10000, 1:20000, 1:30000, 1:40000, 1:50000 or less. In some instances, oligonucleic acids synthesized on the substrate have an average insertion error rate of about 1:500, 1:1000, 1:2000, 1:3000, 1:4000, 1:5000, 1:6000, 1:7000, 1:8000, 1:9000, 1:10000, 1:20000, 1:30000, 1:40000, 1:50000 or less.
  • oligonucleic acids synthesized on the substrate have an average substitution error rate of about 1:500, 1:1000, 1:2000, 1:3000, 1:4000, 1:5000, 1:6000, 1:7000, 1:8000, 1:9000, 1:10000, 1:20000, 1:30000, 1:40000, 1:50000 or less.
  • overall error rate or error rates for individual types of errors such as deletions, insertions, or substitutions for each oligonucleotide synthesized on the substrate, for at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 98%, 99%, 99.5%, or more of the oligonucleotides synthesized on the substrate, or the substrate average may fall between about 1:500 and 1:50000, 1:500 and 1:40000; 1:500 and 1:30000; 1:500 and 1:20000; 1:500 and 1:10000; 1:500 and 1:9000; 1:500 and 1:8000; 1:500 and 1:7000; 1:500 and 1:6000; or 1:500 and 1:5000.
  • the methods and systems described herein for oligonucleic acid synthesis results in minimal synthesis of truncation products that are less than the full length of the predetermined oligonucleic acid sequence.
  • a library of oligonucleic acids are synthesized with less than 1%, 0.5%, 0.1%, 0.05%, 0.01%, 0.005%, 0.001%, or 0.0001% comprising truncation products.
  • the methods and systems described herein for oligonucleic acid synthesis result in minimal synthesis of products that are greater than predetermined oligonucleic acid sequence length.
  • a library of oligonucleic acids are synthesized with less than 1%, 0.5%, 0.1%, 0.05%, 0.01%, 0.005%, 0.001%, or 0.0001% comprising greater than predetermined sequence length.
  • oligonucleic acids synthesized using the systems and methods described herein are optionally evaluated for sequence accuracy prior to subsequent applications, for example, larger nucleic acid assembly.
  • a common method for oligonucleic acid quality control comprises next generation sequencing.
  • Oligonucleic acids synthesized using the methods and substrates described herein are optionally released from the surface from which they were synthesized.
  • oligonucleic acids are cleaved from the surface at this stage.
  • Cleavage may include gas cleavage, e.g., with ammonia or methylamine.
  • all the loci in a single cluster collectively correspond to sequence encoding for a single gene, and optionally, when cleaved, remain on the surface of the loci.
  • the application of ammonia gas simultaneous deprotects phosphates groups protected during the synthesis steps, i.e., removal of electron-withdrawing cyano group.
  • oligonucleic acids are assembled into larger nucleic acids. Synthesized oligonucleic acids are useful, for example, as components for gene assembly/synthesis, site-directed mutagenesis, nucleic acid amplification, microarrays, and sequencing libraries.
  • oligonucleic acids of predetermined sequence are designed to collectively span a large region of a target sequence, such as a gene.
  • larger oligonucleic acids are generated through ligation reactions to join the synthesized oligonucleic acids.
  • a ligation reaction is polymerase chain assembly (PCA).
  • PCA polymerase chain assembly
  • at least of a portion of the oligonucleic acids are designed to include an appended region that is a substrate for universal primer binding.
  • the presynthesized oligonucleic acids include overlaps with each other (e.g., 4, 20, 40 or more bases with overlapping sequence).
  • the oligonucleic acids anneal to complementary fragments and then are filled in by polymerase. Each cycle thus increases the length of various fragments randomly depending on which oligonucleic acids find each other. Complementarity amongst the fragments allows for forming a complete large span of double stranded DNA.
  • an error correction step is conducted using mismatch repair detecting enzymes to remove mismatches in the sequence. Once larger fragments of a target sequence are generated, they can be amplified.
  • a target sequence comprising 5′ and 3′ terminal adaptor sequences is amplified in a polymerase chain reaction (PCR) which includes modified primers, e.g., uracil containing primers the hybridize to the adaptor sequences.
  • PCR polymerase chain reaction
  • modified primers e.g., uracil containing primers the hybridize to the adaptor sequences.
  • modified primers allows for removal of the primers through enzymatic reactions centered on targeting the modified base and/or gaps left by enzymes which cleave the modified base pair from the fragment. What remains is a double stranded amplification product that lacks remnants of adapter sequence. In this way, multiple amplification products can be generated in parallel with the same set of primers to generate different fragments of double stranded DNA.
  • error correction is performed on synthesized oligonucleic acids and/or assembled products.
  • An example strategy for error correction involves site-directed mutagenesis by overlap extension PCR to correct errors, which is optionally coupled with two or more rounds of cloning and sequencing.
  • double-stranded nucleic acids with mismatches, bulges and small loops, chemically altered bases and/or other heteroduplexes are selectively removed from populations of correctly synthesized nucleic acids by affinity purification.
  • error correction is performed using proteins/enzymes that recognize and bind to or next to mismatched or unpaired bases within double stranded nucleic acids to create a single or double strand break or to initiate a strand transfer transposition event.
  • Non-limiting examples of proteins/enzymes for error correction include endonucleases (T7 Endonuclease I, E. coli Endonuclease V, T4 Endonuclease VII, mung bean nuclease, Cell, E. coli Endonuclease IV, UVDE), restriction enzymes, glycosylases, ribonucleases, mismatch repair enzymes, resolvases, helicases, ligases, antibodies specific for mismatches, and their variants.
  • endonucleases T7 Endonuclease I, E. coli Endonuclease V, T4 Endonuclease VII, mung bean nuclease, Cell, E. coli Endonuclease IV, UVDE
  • restriction enzymes glycosylases
  • ribonucleases mismatch repair enzymes
  • resolvases helicases
  • ligases antibodies specific for mismatches, and their
  • error correction enzymes examples include T4 endonuclease 7, T7 endonuclease 1, S1, mung bean endonuclease, MutY, MutS, MutH, MutL, cleavase, CELI, and HINF1.
  • DNA mismatch-binding protein MutS Therms aquaticus
  • error correction is performed using the enzyme Correctase.
  • error correction is performed using SURVEYOR endonuclease (Transgenomic), a mismatch-specific DNA endonuclease that scans for known and unknown mutations and polymorphisms for heteroduplex DNA.
  • a synthesized oligonucleic acid as described herein is amplified in an amplification reaction.
  • a nucleic acid assembled from an oligonucleic acid synthesized by the methods and systems described herein is amplified in an amplification reaction.
  • an amplification reaction includes any method known in the art to amplify one or more nucleic acids.
  • an amplification reaction such as PCR
  • PCR is based on repeated cycles of denaturation, oligonucleic acid primer annealing, and primer extension by thermophilic template dependent polynucleotide polymerase, resulting in the exponential increase in copies of a target nucleic acid sequence flanked by the primers.
  • the two different PCR primers which anneal to opposite strands of the DNA, are positioned so that the polymerase catalyzed extension product of one primer can serve as a template strand for the other, leading to the accumulation of a discrete double stranded fragment whose length is defined by the distance between the 5′ ends of the oligonucleic acid primers.
  • the system comprises the substrate for synthesis support, as described elsewhere herein.
  • the system comprises a device for application of one or more reagents of a synthesis method, for example, an oligonucleic acid synthesizer.
  • the system comprises a device for treating the substrate with a fluid, for example, a flow cell.
  • the system comprises a device for moving the substrate between the application device and the treatment device.
  • an automated system for use with an oligonucleic acid synthesis method described herein that is capable of processing one or more substrates, comprising: a material deposition device for spraying a microdroplet comprising a reagent on a substrate; a scanning transport for scanning the substrate adjacent to the material deposition device to selectively deposit the microdroplet at specified sites; a flow cell for treating the substrate on which the microdroplet is deposited by exposing the substrate to one or more selected fluids; an alignment unit for aligning the substrate correctly relative to the material deposition device each time when the substrate is positioned adjacent to the material deposition device for deposition.
  • the system optionally comprises a treating transport for moving the substrate between the material deposition device and the flow cell for treatment in the flow cell, where the treating transport and said scanning transport are different elements. In other instances, the system does not comprise a treating transport.
  • a device for application of one or more reagents during a synthesis reagent is an oligonucleic acid synthesizer comprising a plurality of material deposition devices.
  • each material deposition device is configured to deposit nucleotide monomers, for example, for phosphoramidite synthesis.
  • the oligonucleic acid synthesizer deposits reagents to the resolved loci, wells, and/or microchannels of a substrate.
  • the oligonucleic acid synthesizer deposits a drop having a diameter less than about 200 um, 100 um, or 50 um in a volume less than about 1000, 500, 100, 50, or 20 pl.
  • the oligonucleic acid synthesizer deposits between about 1 and 10000, 1 and 5000, 100 and 5000, or 1000 and 5000 droplets per second. In some instances, the oligonucleic acid synthesizer uses organic solvents.
  • the substrate is positioned within or sealed within a flow cell.
  • the flow cell provides continuous or discontinuous flow of liquids such as those comprising reagents necessary for reactions within the substrate, for example, oxidizers and/or solvents.
  • the flow cell provides continuous or discontinuous flow of a gas, such as nitrogen, for drying the substrate typically through enhanced evaporation of a volatile substrate.
  • auxiliary devices are useful to improve drying and reduce residual moisture on the surface of the substrate. Examples of such auxiliary drying devices include, without limitation, a vacuum source, depressurizing pump and a vacuum tank.
  • an oligonucleic acid synthesis system comprises one or more flow cells, such as 2, 3, 4, 5, 6, 7, 8, 9, 10, or 20 and one or more substrates, such as 2, 3, 4, 5, 6, 7, 8, 9, 10 or 20.
  • a flow cell is configured to hold and provide reagents to the substrate during one or more steps in a synthesis reaction.
  • a flowcell comprises a lid that slides over the top of a substrate and can be clamped into place to form a pressure tight seal around the edge of the substrate.
  • An adequate seal includes, without limitation, a seal that allows for about 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 atmospheres of pressure.
  • the lid of the flow cell is opened to allow for access to an application device such as an oligonucleic acid synthesizer.
  • an oligonucleic acid synthesizer In some cases, one or more steps of an oligonucleic acid synthesis method are performed on a substrate within a flow cell, without the transport of the substrate.
  • a capping element seals with the substrate, to form a resolved reactor.
  • a substrate having a plurality of clusters is configured to seal with a capping element having a plurality of caps, wherein when the substrate and capping element are sealed, each cluster is separate from another cluster to form separate resolved reactors for each cluster.
  • the capping element is not present in the system or is present and stationary.
  • a resolved reactor is configured to allow for the transfer of fluid, including oligonucleic acids and/or reagents, from the substrate to the capping element and/or vice versa.
  • reactors are interconnected or in fluid communication.
  • Fluid communication of reactors allows for washing and perfusion of new reagents for different steps of a synthesis reaction.
  • the resolved reactors comprise inlets and/or outlets.
  • the inlets and/or outlets are configured for use with a flow cell.
  • a substrate is sealed within a flow cell where reagents can be introduced and flowed through the substrate, after which the reagents are collected.
  • the substrate is drained of fluid and purged with an inert gas such as nitrogen.
  • the flow cell chamber can then be vacuum dried to reduce residual liquids or moisture to less than 1%, 0.1%, 0.01%, 0.001%, 0.0001%, or 0.00001% by volume of the chamber.
  • a vacuum chuck is in fluid communication with the substrate for removing gas.
  • an oligonucleic acid synthesis system comprises one or more elements useful for downstream processing of the synthesized oligonucleic acids.
  • the system comprises a temperature control element such as a thermal cycling device.
  • the temperature control element is used with a plurality of resolved reactors to perform nucleic acid assembly such as PCA and/or nucleic acid amplification such as PCR.
  • any of the systems described herein may be operably linked to a computer and may be automated through a computer either locally or remotely.
  • the methods and systems of the invention may further comprise software programs on computer systems and use thereof.
  • computerized control for the synchronization of the dispense/vacuum/refill functions such as orchestrating and synchronizing the material deposition device movement, dispense action and vacuum actuation are within the bounds of the invention.
  • the computer systems may be programmed to interface between the user specified base sequence and the position of a material deposition device to deliver the correct reagents to specified regions of the substrate.
  • the computer system 1300 illustrated in FIG. 13 may be understood as a logical apparatus that can read instructions from media 1311 and/or a network port 1305 , which can optionally be connected to server 1309 having fixed media 1312 .
  • the system such as shown in FIG. 13 can include a CPU 1301 , disk drives 1303 , optional input devices such as keyboard 1315 and/or mouse 1316 and optional monitor 1307 .
  • Data communication can be achieved through the indicated communication medium to a server at a local or a remote location.
  • the communication medium can include any means of transmitting and/or receiving data.
  • the communication medium can be a network connection, a wireless connection or an internet connection. Such a connection can provide for communication over the World Wide Web. It is envisioned that data relating to the present disclosure can be transmitted over such networks or connections for reception and/or review by a party 1322 as illustrated in FIG. 13 .
  • FIG. 14 is a block diagram illustrating a first example architecture of a computer system 1400 that can be used in connection with example instances of the present invention.
  • the example computer system can include a processor 1402 for processing instructions.
  • processors include: Intel XeonTM processor, AMD OpteronTM processor, Samsung 14-bit RISC ARM 1176JZ(F)-S v1.0TM processor, ARM Cortex-A8 Samsung S5PC100TM processor, ARM Cortex-A8 Apple A4TM processor, Marvell PXA 930TM processor, or a functionally-equivalent processor. Multiple threads of execution can be used for parallel processing. In some instances, multiple processors or processors with multiple cores can also be used, whether in a single computer system, in a cluster, or distributed across systems over a network comprising a plurality of computers, cell phones, and/or personal data assistant devices.
  • a high speed cache 1404 can be connected to, or incorporated in, the processor 1402 to provide a high speed memory for instructions or data that have been recently, or are frequently, used by processor 1402 .
  • the processor 1402 is connected to a north bridge 1406 by a processor bus 1408 .
  • the north bridge 1406 is connected to random access memory (RAM) 1410 by a memory bus 1412 and manages access to the RAM 1410 by the processor 1402 .
  • the north bridge 1406 is also connected to a south bridge 1414 by a chipset bus 1416 .
  • the south bridge 1414 is, in turn, connected to a peripheral bus 1418 .
  • the peripheral bus can be, for example, PCI, PCI-X, PCI Express, or other peripheral bus.
  • the north bridge and south bridge are often referred to as a processor chipset and manage data transfer between the processor, RAM, and peripheral components on the peripheral bus 1418 .
  • the functionality of the north bridge can be incorporated into the processor instead of using a separate north bridge chip.
  • system 1400 can include an accelerator card 1422 attached to the peripheral bus 1418 .
  • the accelerator can include field programmable gate arrays (FPGAs) or other hardware for accelerating certain processing.
  • FPGAs field programmable gate arrays
  • an accelerator can be used for adaptive data restructuring or to evaluate algebraic expressions used in extended set processing.
  • the system 1400 includes an operating system for managing system resources; non-limiting examples of operating systems include: Linux, WindowsTM, MACOSTM, BlackBerry OSTM, iOSTM, and other functionally-equivalent operating systems, as well as application software running on top of the operating system for managing data storage and optimization in accordance with example instances of the present invention.
  • system 1400 also includes network interface cards (NICs) 1420 and 1421 connected to the peripheral bus for providing network interfaces to external storage, such as Network Attached Storage (NAS) and other computer systems that can be used for distributed parallel processing.
  • NICs network interface cards
  • NAS Network Attached Storage
  • FIG. 15 is a diagram showing a network 1500 with a plurality of computer systems 1502 a , and 1502 b , a plurality of cell phones and personal data assistants 1502 c , and Network Attached Storage (NAS) 1504 a , and 1504 b .
  • systems 1502 a , 1502 b , and 1502 c can manage data storage and optimize data access for data stored in Network Attached Storage (NAS) 1504 a and 1504 b .
  • a mathematical model can be used for the data and be evaluated using distributed parallel processing across computer systems 1502 a , and 1502 b , and cell phone and personal data assistant systems 1502 c .
  • Computer systems 1502 a , and 1502 b , and cell phone and personal data assistant systems 1502 c can also provide parallel processing for adaptive data restructuring of the data stored in Network Attached Storage (NAS) 1504 a and 1504 b .
  • FIG. 15 illustrates an example only, and a wide variety of other computer architectures and systems can be used in conjunction with the various instances of the present invention.
  • a blade server can be used to provide parallel processing.
  • Processor blades can be connected through a back plane to provide parallel processing.
  • Storage can also be connected to the back plane or as Network Attached Storage (NAS) through a separate network interface.
  • NAS Network Attached Storage
  • processors can maintain separate memory spaces and transmit data through network interfaces, back plane or other connectors for parallel processing by other processors. In other instances, some or all of the processors can use a shared virtual address memory space.
  • FIG. 16 is a block diagram of a multiprocessor computer system 1600 using a shared virtual address memory space in accordance with an example embodiment.
  • the system includes a plurality of processors 1602 a - f that can access a shared memory subsystem 1604 .
  • the system incorporates a plurality of programmable hardware memory algorithm processors (MAPs) 1606 a - f in the memory subsystem 1604 .
  • MAPs programmable hardware memory algorithm processors
  • Each MAP 1606 a - f can comprise a memory 1608 a - f and one or more field programmable gate arrays (FPGAs) 1610 a - f
  • the MAP provides a configurable functional unit and particular algorithms or portions of algorithms can be provided to the FPGAs 1610 a - f for processing in close coordination with a respective processor.
  • the MAPs can be used to evaluate algebraic expressions regarding the data model and to perform adaptive data restructuring in example instances.
  • each MAP is globally accessible by all of the processors for these purposes.
  • each MAP can use Direct Memory Access (DMA) to access an associated memory 1608 a - f , allowing it to execute tasks independently of, and asynchronously from the respective microprocessor 1602 a - f .
  • DMA Direct Memory Access
  • a MAP can feed results directly to another MAP for pipelining and parallel execution of algorithms.
  • the above computer architectures and systems are examples only, and a wide variety of other computer, cell phone, and personal data assistant architectures and systems can be used in connection with example instances, including systems using any combination of general processors, co-processors, FPGAs and other programmable logic devices, system on chips (SOCs), application specific integrated circuits (ASICs), and other processing and logic elements.
  • SOCs system on chips
  • ASICs application specific integrated circuits
  • all or part of the computer system can be implemented in software or hardware.
  • Any variety of data storage media can be used in connection with example instances, including random access memory, hard drives, flash memory, tape drives, disk arrays, Network Attached Storage (NAS) and other local or distributed data storage devices and systems.
  • NAS Network Attached Storage
  • the computer system can be implemented using software modules executing on any of the above or other computer architectures and systems.
  • the functions of the system can be implemented partially or completely in firmware, programmable logic devices such as field programmable gate arrays (FPGAs) as referenced in FIG. 16 , system on chips (SOCs), application specific integrated circuits (ASICs), or other processing and logic elements.
  • FPGAs field programmable gate arrays
  • SOCs system on chips
  • ASICs application specific integrated circuits
  • the Set Processor and Optimizer can be implemented with hardware acceleration through the use of a hardware accelerator card, such as accelerator card 1322 illustrated in FIG. 13 .
  • Example 1 Functionalization of a Substrate Surface
  • a substrate was functionalized to support the attachment and synthesis of a library of oligonucleic acids.
  • the substrate surface was first wet cleaned using a piranha solution comprising 90% H 2 SO 4 and 10% H 2 O 2 for 20 minutes.
  • the substrate was rinsed in several beakers with DI water, held under a DI water gooseneck faucet for 5 min, and dried with N 2 .
  • the substrate was subsequently soaked in NH 4 OH (1:100; 3 mL:300 mL) for 5 min, rinsed with DI water using a handgun, soaked in three successive beakers with DI water for 1 min each, and then rinsed again with DI water using the handgun.
  • the substrate was then plasma cleaned by exposing the substrate surface to O 2 .
  • a SAMCO PC-300 instrument was used to plasma etch O 2 at 250 watts for 1 min in downstream mode.
  • the cleaned substrate surface was actively functionalized with a solution comprising N-(3-triethoxysilylpropyl)-4-hydroxybutyramide using a YES-1224P vapor deposition oven system with the following parameters: 0.5 to 1 Torr, 60 min, 70° C., 135° C. vaporizer.
  • the substrate surface was resist coated using a Brewer Science 200 ⁇ spin coater.
  • SPRTM 3612 photoresist was spin coated on the substrate at 2500 rpm for 40 sec.
  • the substrate was pre-baked for 30 min at 90° C. on a Brewer hot plate.
  • the substrate was subjected to photolithography using a Karl Suss MA6 mask aligner instrument.
  • the substrate was exposed for 2.2 sec and developed for 1 min in MSF 26A. Remaining developer was rinsed with the handgun and the substrate soaked in water for 5 min.
  • the substrate was baked for 30 min at 100° C. in the oven, followed by visual inspection for lithography defects using a Nikon L200.
  • a plasma cleaning process was used to remove residual resist using the SAMCO PC-300 instrument to O 2 plasma etch at 250 watts for 1 min.
  • the substrate surface was passively functionalized with a 100 ⁇ L solution of perfluorooctyltrichlorosilane mixed with 10 ⁇ L light mineral oil.
  • the substrate was placed in a chamber, pumped for 10 min, and then the valve was closed to the pump and left to stand for 10 min. The chamber was vented to air.
  • the substrate was resist stripped by performing two soaks for 5 min in 500 mL NMP at 70° C. with ultrasonication at maximum power (9 on Crest system). The substrate was then soaked for 5 min in 500 mL isopropanol at room temperature with ultrasonication at maximum power.
  • the substrate was dipped in 300 mL of 200 proof ethanol and blown dry with N 2 .
  • the functionalized surface was activated to serve as a support for oligonucleic acid synthesis.
  • Substrates were manufactured to comprise a plurality of clusters each comprising a plurality of distinct loci configured to provide structural support for oligonucleic acid synthesis.
  • Substrate starting material was a 200 mm standard, double-sided polished silicon wafer having a 725 um thickness.
  • Substrates were processed by a method comprising thermal oxidation at 1000 ⁇ , photolithography using a Karl Suss MA6 mask aligner to generate fiducial structures; oxide etching down to the silicon; and resist stripping.
  • Prepared substrates have 6,144 clusters, with each cluster having 121 reaction sites or loci for oligonucleic acid synthesis.
  • the clusters are organized into 24 sub-fields, which each comprise a 16 ⁇ 16 array of clusters.
  • FIGS. 1 - 3 A schematic of a substrate produced is shown in FIGS. 1 - 3 .
  • the substrate has a dimension of 140.000 mm by 90.000 mm.
  • the vertical distance between the centers of two adjacent clusters in one substrate is 1079.210 um and in another substrate 1142.694 um.
  • the horizontal distance between the centers of two adjacent clusters in the substrate is 1125.0 um.
  • An expanded view of a cluster of the substrate is shown in FIG. 3 .
  • Each cluster has 121 loci, which are separated so that the horizontal distance between two adjacent loci is 75.000 um and the vertical distance between two loci is 63.483 um.
  • the horizontal distance between the edges of two adjacent loci is 24.0 um.
  • Substrates manufactured in this example were processed to generate three-dimensional loci having shapes configured to increase surface area to volume. Examples of locus shapes prepared using the methods described in this example are shown in FIG. 5 .
  • a Silicon on Insulator (SOI) silicon wafer (sub-field size of 32.00 ⁇ 32.00 mm) was oxidized, and the device side processed by photolithography, deep RIE and photoresist stripping. The handle side of the substrate was processed by photolithography, deep RIE, photoresist stripping, and etching by removal of oxide layer (BOX etch).
  • SOI Silicon on Insulator
  • the processed substrate has a plurality of wells or holes within the handle layer, each having a width of 1.150 mm, wherein each channel has a plurality of microchannels having shapes that allow for an increase in surface area to volume.
  • the smallest etch size for a feature of a shape of a microchannel within a substrate prepared in this example was 5 um.
  • the distance between the centers of two adjacent clusters (wells) is 1.693 mm in all directions.
  • the distance between the centers of two adjacent loci (microchannels) is 97.765 in a horizontal direction and 84.667 um in a vertical direction.
  • the prepared substrate has a set of markings or fiducials of 0.5 mm diameter.
  • the width of the main channel is 1.150 mm and the width of the microchannel is 5 um. Detailed features of substrates prepared using these methods are shown in FIGS. 5 - 8 .
  • a cluster of a processed substrate having double comb shaped loci is shown in a bird's eye view in FIG. 6 .
  • the combined height of the two longest teeth is 57 um.
  • the distance between two teeth of the comb is 14.0 um.
  • the width of the comb handle is 5 um.
  • the combined height of the two shortest teeth is 38.0 um.
  • the width of the comb in the horizontal direction is 47.0 urn.
  • a cluster of a processed substrate having single comb shaped loci is shown in a bird's eye view in FIG. 7 .
  • the height of the longest tooth is 49.0 um.
  • the distance between two teeth of the comb is 14.0 um.
  • the width of the comb handle is 5 um.
  • the height of the shortest tooth is 39.0 um.
  • the width of the comb in the horizontal direction is 47.0 um.
  • a cluster of a processed substrate having serpentine shaped loci is shown in a bird's eye view in FIG. 8 .
  • the height of the loci shape is 54 um.
  • the distance between two lines of the shape is 14 um.
  • the width of a line of the shape is 5 um.
  • the surface and volume parameters of loci having high surface area shapes (double comb, single comb, and serpentine) prepared using these methods were compared with the parameters of a locus having a revolver shape (barrel comprising 5 channels). The comparison is shown in Table 4.
  • the loci having a comb or serpentine shape had a lower substrate volume than a substrate having revolver loci.
  • the loci having a comb or serpentine shape had a greater surface area than a substrate having revolver loci.
  • the loci having a comb or serpentine shape had a greater surface area to volume ratio than a substrate having revolver loci.
  • a substantially planar substrate functionalized for oligonucleic acid synthesis was assembled into a flow cell and connected to an Applied Biosystems ABI394 DNA Synthesizer.
  • the substrate was uniformly functionalized with N-(3-triethoxysilylpropyl)-4-hydroxybutyramide.
  • the substrate was functionalized with a 5/95 mix of 11-acetoxyundecyltriethoxysilane and N-decyltriethoxysilane.
  • oligonucleic acids were extracted from the substrate surface and analyzed on a BioAnalyzer chip. Oligonucleic acid products were PCR amplified, cloned and Sanger sequenced. Table 6 summarizes the Sanger sequencing results for samples taken from spots 1-5 from one chip and spots 6-10 from a second chip.
  • Table 7 summarizes key error characteristics for the sequences obtained from the oligonucleic acid samples from spots 1-10.
  • PCA reaction mixture drops of about 400 nL were dispensed using a Mantis dispenser (Formulatrix, MA) on the top of channels of a device side of a three-dimensional substrate having a plurality of loci microchannels in fluid communication with a single main channel of a cluster.
  • a nanoreactor chip was manually mated with the substrate to pick up the droplets having the PCA reaction mixture and oligonucleic acids from each channel.
  • the droplets were picked up into individual nanoreactors in the nanoreactor chip by releasing the nanoreactor from the substrate immediately after pick up.
  • the nanoreactors were sealed with a heat sealing film, placed in a thermocycler for PCA.
  • PCA thermocycling conditions are shown in Table 10.
  • a PCA reaction performed in plastic tube was also ran on the gel as a positive control (panel 11 ), which shows a product having a similar size to the products from wells 1 - 10 .
  • a negative control (panel 12 ) was also run on the gel, which corresponds to a PCR reaction ran without a PCA template. The BioAnalyzer data is not shown.
  • a gene of about 1 kbp (SEQ ID.: 67; Table 13) was assembled using 6 purchased Ultramer oligonucleotides (SEQ ID NO.: 68-73; Table 13) and assembled in a PCA reaction. Ultramer oligonucleotides are expected to have error rates of at least 1 in 500 to 1 in 200 nucleotides.
  • the assembled gene was amplified by PCR using a forward primer (5′ ATGACCATGATTACGGATTCACTGGCC3′ SEQ ID NO.: 65) and a reverse primer (5′GATAGAGATTCGGGATTTCGGCGCTCC3′ SEQ ID NO.: 66). The amplified assembled gene was analyzed in a BioAnalyzer and cloned.
  • DNA preparations from 24 colonies were Sanger sequenced.
  • the BioAnalyzer analysis provided a broad peak and a tail for the uncorrected gene, indicating a high error rate.
  • the sequencing indicated an error rate of 1/789 bases.
  • Two rounds of error correction were followed using CorectASE (Life Technologies) according to the manufacturer's instructions.
  • the resulting gene samples were analyzed in a BioAnalyzer after round one and round two and cloned. Twenty-four colonies were picked for sequencing.
  • the sequencing results indicated an error rate of 1/5190 bases and 1/6315 bases after the first and second rounds of error correction, respectively.
  • a substrate 1700 comprising 256 clusters each comprising 121 loci on a flat silicon plate was manufactured as shown in FIG. 17 .
  • An expanded view of a cluster is shown in 1705 with 121 loci.
  • Loci from 240 of the 256 clusters provided an attachment and support for the synthesis of oligonucleic acids having distinct sequences. Oligonucleic acid synthesis was performed by phosphoramidite chemistry using general methods from Table 5 in Example 4. Loci from 16 of the 256 clusters were control clusters.
  • the distribution of the 29,040 unique oligonucleic acids synthesized (240 ⁇ 121) is shown in FIG. 18 .
  • the distribution of unique oligonucleic acids synthesized in 4 representative clusters is shown in FIG. 19 .
  • the error rate for each oligonucleic acid was determined using an Illumina MiSeq gene sequencer.
  • the error rate distribution for the 29,040 unique oligonucleic acids is shown in FIG. 20 and averages around 1 in 500 bases, with some error rates as low as 1 in 800 bases.
  • the error rate distribution for unique oligonucleic acids in four representative clusters is shown in FIG. 21 .
  • the library of 29,040 unique oligonucleic acids was synthesized in less than 20 hours.
  • Oligonucleic acids synthesized within the loci of a cluster had overlapping sequences with one another so that when all oligonucleic acids synthesized within one cluster are pooled, they are ready for assembly into a larger nucleic acid or gene using PCA. Oligonucleic acids within a cluster were pooled and assembled using PCA reaction conditions similar to those described in Example 5. The 240 unique genes were synthesized in 3 business days, however, with 24 hour a day operation, the 240 unique genes could be synthesized in less time. The assembled genes from each of the 240 clusters were sequenced using an Illumina MiSeq gene sequencer. The read counts for the assembled genes are represented in FIG. 22 . The assembled gene products were visualized on a DNA gel as shown in FIGS. 23 - 26 . The genes synthesized ranged in size from 838 base pairs to 1470 base pairs. All 240 gene products were generated with their expected size. An output from the sequencing run is shown in FIG. 27 .
  • a substrate comprising three-dimensional features with high surface area loci was manufactured according to the methods similar to that of Example 3. Each locus was manufactured to have a single comb shape.
  • the substrate has a plurality of clusters corresponding to a plurality of wells, wherein each channel is 1.150 mm in diameter and includes 109 loci in the form of microchannels.
  • FIG. 10 C provides a depiction for a collection of microchannels/loci extending from a main channel.
  • Microchannels of the substrate were functionalized and used as an attachment and support for the synthesis of distinct oligonucleic acids.
  • a library of oligonucleic acids was synthesized on the substrate and subsequently gas cleaved from the surface for sequence analysis using an Illumina MiSeq.
  • Error mismatch rates for oligonucleic acids synthesized within each cluster were calculated (data not shown). Error rates were from 1 in 7,000 bases to as high as 1 in 100 bases. The average mismatch error rate was 1 in 586.82 bases.
  • Error deletion rates for oligonucleic acids synthesized within each cluster were calculated (data not shown). Error rates were from around 1 in 1,000 bases to around 1 in 2,000 bases. The average deletion error rate was 1 in 1,966.86 bases.
  • Insertion rates for oligonucleic acids synthesized within each cluster were calculated (data not shown). Error rates were from 1 in 2,700 bases to around 1 in 10,000 bases. The average insertion error rate was 1 in 4,740.03 bases.
  • the percentage of perfect oligonucleic acids synthesized (a perfect sequence being 100% identical to a preselected nucleic acid sequence) within each cluster were calculated (data not shown). The percentage of perfect sequences ranged from about 70% to about 93.54% perfect sequences. Overall more than 90% of the oligonucleic acids had perfect sequences.
  • oligonucleic acid library was synthesized as described in Example 8. Each oligonucleic acid synthesized on a locus of a cluster was tethered to the locus by a linker. Error rate as a function of base distance from substrate surface was analyzed and graphed as depicted in FIG. 28 . The lowest error rates correspond to oligonucleic acids with tether between about 12 and 25 bases from the surface.
  • Oligonucleic acids of various sequences and lengths were synthesized by phosphoramidite chemistry using methods as generally described in Table 5 of Example 4. Oligonucleic acids having lengths from 25 bases to 200 bases were synthesized within different clusters of a substrate. The synthesized oligonucleic acids were released from the surface, collected, and visualized by gel electrophoresis.
  • FIG. 29 provides a captured image of the electrophoresis gel loaded with representative synthesized oligonucleic acids having lengths of 25, 50, 75, 100, 125, 150, 175 and 200 nucleotides.
  • the methods and substrates described herein allow for the simultaneous synthesis of a plurality of oligonucleic acids each having different sequences and, in some cases, different sequence lengths.
  • oligonucleic acids having 200 bases were synthesized on, and removed from a substrate. These synthesized oligonucleic acids were released from the substrate and used in downstream processes, such as visualization by gel electrophoresis.
  • Representative quantities of synthesized oligonucleic acids extracted from each cluster in this example ranged from 113 fmol to 344 fmol.
  • Representative yields from each cluster ranged from 48 pmol/cm 2 to 145 pmol/cm 2 .

Abstract

Devices and methods for de novo synthesis of large and highly accurate libraries of oligonucleic acids are provided herein. Devices include structures having a main channel and microchannels, where the microchannels have a high surface area to volume ratio. Devices disclosed herein provide for de novo synthesis of oligonucleic acids having a low error rate.

Description

    CROSS REFERENCE
  • This application is a continuation of U.S. patent application Ser. No. 16/921,712, filed Jul. 6, 2020, which is a continuation of U.S. patent application Ser. No. 15/960,319, filed on Apr. 23, 2018, now U.S. Pat. No. 10,744,477 issued Aug. 18, 2020, which is a continuation of U.S. patent application Ser. No. 15/135,434 filed Apr. 21, 2016, now U.S. Pat. No. 9,981,239 issued May 29, 2018, which claims the benefit of U.S. Provisional Application No. 62/150,795 filed Apr. 21, 2015, and U.S. Provisional Application No. 62/220,856 filed Sep. 18, 2015, each of which are herein incorporated by reference in their entirety.
  • SEQUENCE LISTING
  • The instant application contains a Sequence Listing which has been submitted electronically in .XML format and is hereby incorporated by reference in its entirety. Said .XML copy, created on May 17, 2023, is named 44854_713_303_SL.xml and is 105,152 bytes in size.
  • BACKGROUND
  • Highly efficient chemical gene synthesis with high fidelity and low cost has a central role in biotechnology and medicine, and in basic biomedical research. De novo gene synthesis is a powerful tool for basic biological research and biotechnology applications. While various methods are known for the synthesis of relatively short fragments in a small scale, these techniques often suffer from scalability, automation, speed, accuracy, and cost.
  • BRIEF SUMMARY
  • Provided herein are devices for synthesizing oligonucleic acids, comprising: a plate; a main channel, wherein the main channel extends vertically into the plate from an opening on a top side of the plate, and wherein the main channel has a width of 0.5 to 2 mm; and a plurality of microchannels connected to the main channel, wherein each microchannel of the plurality of microchannels extends vertically from an opening on a bottom side of the plate into the main channel, and wherein each microchannel of the plurality of microchannels has a surface area to volume ratio of greater than 0.2 (1/um). Devices are further provided wherein the surface area to volume ratio provides for rapid exchange of chemical exposure during de novo synthesis of oligonucleic acids. Devices are further provided wherein the surface area to volume ratio is about 0.4 (1/um). Devices are further provided wherein the surface area to volume ratio is greater than 0.4 (1/um). Devices are further provided wherein the surface area to volume ratio is 0.41 (1/um). Devices are further provided wherein each microchannel of the plurality of microchannels has a surface area greater than 10,000 um2. Devices are further provided wherein each microchannel of the plurality of microchannels has a surface area greater than 12,000 um2. Devices are further provided wherein each microchannel of the plurality of microchannels has a surface area of about 13,000 um2. Devices are further provided wherein the plurality of microchannels comprises 50 to 500 microchannels. Devices are further provided wherein the plurality of microchannels comprises 100 to 150 microchannels. Devices are further provided wherein a ratio of width to depth of a narrowest segment of each microchannel is from 0.5 to 0.01. Devices are further provided wherein a ratio of width to depth of a narrowest segment of each microchannel is about 0.05, 0.1, or 0.2. Devices are further provided wherein each microchannel of the plurality of microchannels has a total width of 30 um to 100 um. Devices are further provided wherein each microchannel of the plurality of microchannels has a total width of about 60 um. Devices are further provided wherein each microchannel of the plurality of microchannels has a depth of 10 to 500 um. Devices are further provided wherein each microchannel of the plurality of microchannels has a depth of about 30 um. Devices are further provided wherein the main channel has a width from 0.5 to 1.5 mm. Devices are further provided wherein the main channel has a width of about 1.2 mm. Devices are further provided wherein the main channel has a width of 1.15 mm. Devices are further provided wherein the device comprises more than 250 main channels. Devices are further provided wherein the device comprises more than 10,000 main channels. Devices are further provided that further comprising a first molecule, wherein the first molecule is bound to an interior surface of the plurality of microchannels and comprises a reactive group that binds to a nucleoside phosphoramidite. Devices are further provided wherein the first molecule is a silane. Devices are further provided wherein the first molecule is an aminosilane. Devices are further provided wherein the first molecule is 11-acetoxyundecyltriethoxysilane, n-decyltriethoxysilane, (3-aminopropyl)trimethoxysilane, (3-aminopropyl)triethoxysilane, glycidyloxypropyl/trimethoxysilane or N-(3-triethoxysilylpropyl)-4-hydroxybutyramide. Devices are further provided further comprising a second molecule, wherein the second molecule is bound to an interior surface of the main channel and lacks a reactive group that binds to a nucleoside phosphoramidite. Devices are further provided wherein the second molecule is a fluorosilane. Devices are further provided wherein the fluorosilane is (tridecafluorotetrahydrooctyl)-triethoxysilane. Devices are further provided wherein the first molecule has a higher surface energy than the second molecule, and wherein the first molecule has a greater hydrophobicity than the second molecule. Devices are further provided wherein the plate is a silicon plate. Devices are further provided wherein the device comprises at least 30,000 microchannels. Devices are further provided wherein the device comprises at least 700,000 microchannels.
  • Provided herein are devices for synthesizing oligonucleic acids, comprising: a plate; a main channel, wherein the main channel extends vertically into the silicon plate from an opening on a top side of the plate, and wherein the main channel has a width of 0.5 to 2 mm; and a plurality of microchannels connected to the main channel, wherein each microchannel of the plurality of microchannels extends vertically from an opening on a bottom side of the plate into the main channel, and wherein each microchannel of the plurality of microchannels comprises a total width that is less than 100 um, a microchannel surface area greater than 10,000 um, and a maximum width for the narrowest segment of the microchannel of 10 um. Devices are further provided wherein each microchannel of the plurality of microchannels has a surface area greater than 12,000 um2. Devices are further provided wherein each microchannel of the plurality of microchannels has a surface area of about 13,000 um2. Devices are further provided wherein the plurality of microchannels comprises 50 to 500 microchannels. Devices are further provided wherein the plurality of microchannels comprises 100 to 150 microchannels. Devices are further provided wherein a ratio of width to depth of a narrowest segment of each microchannel is from 0.5 to 0.01. Devices are further provided wherein a ratio of width to depth of a narrowest segment of each microchannel is about 0.05, 0.1, or 0.2. Devices are further provided wherein each microchannel of the plurality of microchannels has a total width of 30 um to 100 um. Devices are further provided wherein each microchannel of the plurality of microchannels has a total width of about 60 um. Devices are further provided wherein each microchannel of the plurality of microchannels has a depth of 10 to 500 um. Devices are further provided wherein each microchannel of the plurality of microchannels has a depth of about 30 um. Devices are further provided wherein the main channel has a width from 0.5 to 1.5 mm. Devices are further provided wherein the main channel has a width of about 1.2 mm. Devices are further provided wherein the main channel has a width of 1.15 mm. Devices are further provided wherein the device comprises more than 250 main channels. Devices are further provided wherein the device comprises more than 10,000 main channels. Devices are further provided further comprising a first molecule, wherein the first molecule is bound to an interior surface of the plurality of microchannels and comprises a reactive group that binds to a nucleoside phosphoramidite. Devices are further provided wherein the first molecule is a silane. Devices are further provided wherein the first molecule is an aminosilane. Devices are further provided wherein the first molecule is 11-acetoxyundecyltriethoxysilane, n-decyltriethoxysilane, (3-aminopropyl)trimethoxysilane, (3-aminopropyl)triethoxysilane, glycidyloxypropyl/trimethoxysilane or N-(3-triethoxysilylpropyl)-4-hydroxybutyramide. Devices are further provided further comprising a second molecule, wherein the second molecule is bound to an interior surface of the main channel and lacks a reactive group that binds to a nucleoside phosphoramidite. Devices are further provided wherein the second molecule is a fluorosilane. Devices are further provided wherein the fluorosilane is (tridecafluorotetrahydrooctyl)-triethoxysilane. Devices are further provided wherein the first molecule has a higher surface energy than the second molecule, and wherein the first molecule has a greater hydrophobicity than the second molecule. Devices are further provided wherein the plate is a silicon plate. Devices are further provided wherein the device comprises at least 30,000 microchannels. Devices are further provided wherein the device comprises at least 700,000 microchannels.
  • Provided herein are methods for de novo oligonucleic acid synthesis, comprising: providing predetermined sequences for a plurality of non-identical oligonucleic acids; providing a device described herein; adding a droplet of fluid comprising an extension reaction reagent specific to a microchannel; allowing sufficient time for an extension reaction step to occur; and repeating steps (c) and (d) until the plurality of non-identical oligonucleic acids are synthesized, wherein each oligonucleic acid at least 10 bases in length and attached to an inside region of the microchannel, and wherein the synthesized non-identical oligonucleic acids encode sequences with an aggregate error rate of less than 1 in 2000 bases compared to the predetermined sequences. Methods are further provided wherein the synthesized non-identical oligonucleic acids encode sequences with an aggregate error rate of less than 1 in 3000 bases compared to the predetermined sequences. Methods are further provided wherein the synthesized non-identical oligonucleic acids encode sequences with an insertion error rate of less than 1 in 5000 bases compared to the predetermined sequences. Methods are further provided wherein the synthesized non-identical oligonucleic acids encode sequences with a deletion error rate of less than 1 in 2000 bases compared to the predetermined sequences. Methods are further provided that further comprising washing the surface with a washing reagent, and wherein washing removes greater than 95% of unincorporated extension reaction reagent. Methods are further provided wherein washing removes greater than 99% of unincorporated extension reaction reagent. Methods are further provided wherein the droplet of fluid is less than about 32 pL in volume. Methods are further provided wherein the method is completed in less than 24 hours. Methods are further provided wherein the synthesized plurality of non-identical oligonucleic acids are fluidically connected to a single main channel and collectively encode for a single gene. Methods are further provided wherein the oligonucleic acids collectively encode for at least 200 genes at least 1 kb in length. Methods are further provided further comprising: releasing the plurality of non-identical oligonucleic acids from the surface; and subjecting the plurality of non-identical oligonucleic acids to a polymerase chain assembly reaction to assemble at least 200 genes. Methods are further provided wherein the at least 200 genes have an aggregate error rate of less than 1 in 2000 bases compared to the predetermined sequences without correcting errors. Methods are further provided wherein the at least 200 genes have an aggregate error rate of less than 1 in 3000 bases compared to the predetermined sequences without correcting errors. Methods are further provided wherein each oligonucleic acid has a tether region 12 to 25 bases in length. Methods are further provided wherein the tether region is homopolymeric. Methods are further provided wherein each oligonucleic acid is at least 30 bases in length. Methods are further provided wherein each oligonucleic acid 50 to 500 bases in length.
  • Provided herein are devices for synthesizing oligonucleic acids, comprising a silicon plate; a main channel, wherein the main channel extends vertically into the silicon plate from an opening on a top side of the silicon plate, and wherein the main channel has a width of 0.5 to 2 mm; and 50 to 500 microchannels connected to the main channel, wherein each of the 50 to 500 microchannels extends vertically from an opening on a bottom side of the silicon plate into the main channel, and wherein each microchannel of the 50 to 500 microchannels has a surface area to volume ratio of greater than about 0.4 (1/um), and a ratio of width to depth of a narrowest segment of each microchannel is from 0.5 to 0.01.
  • INCORPORATION BY REFERENCE
  • All publications, patents, and patent applications disclosed herein are incorporated by reference to the same extent as if each individual publication, patent, or patent application was specifically and individually indicated to be incorporated by reference. In the event of a conflict between a term disclosed herein and a term in an incorporated reference, the term herein controls.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 illustrates a plate configured for oligonucleic acid synthesis comprising 24 regions, or sub-fields, each having an array of 256 clusters.
  • FIG. 2 illustrates a closer view of the sub-field in FIG. 1 having 16×16 of clusters, each cluster having 121 individual loci.
  • FIG. 3 illustrates a detailed view of the cluster in FIG. 2 , where the cluster has 121 loci.
  • FIG. 4A illustrates a front view of a plate with a plurality of microchannels.
  • FIG. 4B illustrates a sectional view of plate with a plurality of microchannels.
  • FIG. 5 illustrates three-dimensional arrangements for microchannels.
  • FIG. 6 illustrates a cluster having a plurality of loci with double comb shapes.
  • FIG. 7 illustrates a cluster having a plurality of loci with single comb shapes.
  • FIG. 8 illustrates a cluster having a plurality of loci with single serpentine shapes.
  • FIGS. 9A-9F illustrate a workflow for the passive and active functionalization of an etched substrate.
  • FIGS. 10A-10C illustrate reagent deposition directly into microchannels within a main channel, where: microchannels are actively functionalized (FIG. 10A), main channels are passively functionalized (FIG. 10B); and microchannels are actively functionalized and main channels are passively functionalized (FIG. 10C). The dotted lines indicate that the image depicts one main channel of many in a single substrate (e.g., a plate).
  • FIGS. 11A-11C illustrate reagent deposition directly into microchannels within a main channel, where a plate contains a silicon oxide later at a boundary between a main channel and a microchannel, where: microchannels are actively functionalized (FIG. 11A), main channels are passively functionalized (FIG. 11B); and microchannels are actively functionalized and main channels are passively functionalized (FIG. 11C). The dotted lines indicate that the image depicts one main channel of many in a single substrate (e.g., a plate).
  • FIG. 12 illustrates a workflow for de novo oligonucleotide synthesis.
  • FIG. 13 illustrates a computer system.
  • FIG. 14 is a block diagram illustrating architecture of a computer system.
  • FIG. 15 is a diagram demonstrating a network configured to incorporate a plurality of computer systems, a plurality of cell phones and personal data assistants, and Network Attached Storage (NAS).
  • FIG. 16 is a block diagram of a multiprocessor computer system using a shared virtual address memory space.
  • FIG. 17 depicts read counts from a sub-array having 256 clusters (left), and an image of a cluster having 121 loci (right).
  • FIG. 18 is a graphical representation of oligonucleic acid frequency versus abundance from an experiment where oligonucleic acids were synthesized on the substrate of FIG. 17 .
  • FIG. 19 is a graphical representation of oligonucleic acid frequency versus abundance for four representative clusters of the substrate of FIG. 17 .
  • FIG. 20 is a graphical representation of oligonucleic acid frequency versus error rate for oligonucleic acids synthesized on the substrate of FIG. 17 .
  • FIG. 21 is a graphical representation of oligonucleic acid frequency versus error rate for oligonucleic acids synthesized on four representative clusters of the substrate of FIG. 17 .
  • FIG. 22 is a representation of read counts from 240 assembled genes from a library of oligonucleic acids synthesized on a substrate.
  • FIGS. 23, 24, 25 and 26 provide digital images from gel electrophoresis of 240 assembled genes from a library of oligonucleic acids synthesized on the substrate of FIG. 17 .
  • FIG. 27 provides an output reading from next generation sequencing of 240 assembled genes from a library of oligonucleic acids synthesized on the substrate of FIG. 17 .
  • FIG. 28 provides a graphical representation of insertion/deletion (“indel”) error count as a function of read cycle for a synthesized library of oligonucleic acids.
  • FIG. 29 provides a digital image of an electrophoresis gel showing 25mer to 200mer oligonucleic acids synthesized using a substrate and methods provided herein.
  • DETAILED DESCRIPTION
  • The present disclosure provides compositions, devices, methods and systems for the de novo synthesis of a library of oligonucleic acids with low error rates. The oligonucleic acids are useful components, such as for the generation of larger nucleic acids, such as genes as part of gene libraries.
  • Described herein are devices having structural features that control the flow of fluid through small channels (“microchannels”) which also serve as locations for oligonucleic acid extension. Factors that can impact the flow of fluid throw the surface include, without limitation, the number of microchannels, microchannel size, the shape of the microchannels, the width of a main channel which a group of microchannels collectively connect, and the chemical properties of surfaces involved (e.g., hydrophobicity and surface energy). During oligonucleic acid synthesis, it is desirable to have channels to have enough width to support the extension of multiple oligonucleic acids, while at the same time be narrow enough to support rapid fluid transfer. Rapid fluid transfer is desirable to provide for efficient chemical exchange during various steps of the de novo nucleic acid synthesis process, and reduce unwanted side reactions that may lead to increased error rates. Devices described herein increase fluid transfer rate through a substrate and also increase the amount of surface available for nucleic acid extension is by having microchannels with a high surface area to volume ratio. Such devices also provide for synthesis of oligonucleic acids with low error rates.
  • In some cases, oligonucleic acids synthesized within a cluster of extension locations (“loci”) comprise specific predetermined sequences that are configured to be assembled to generate a larger nucleic acid. In this manner, the parallel generation of genes is done on a single substrate. The average error rates for oligonucleic acids synthesized within a library using the systems and methods provided are often less than 1 in 1000, and are preferably less than about 1 in 2000, 1 in 5000 or less often.
  • Definitions
  • The present disclosure employs, unless otherwise indicated, conventional molecular biology techniques, which are within the skill of the art. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as is commonly understood by one of ordinary skill in the art to which these inventions belong.
  • Throughout this disclosure, various instances are presented in a range format. It should be understood that the description in range format is merely for convenience and brevity and should not be construed as an inflexible limitation on the scope of any instances. Accordingly, the description of a range should be considered to have specifically described all the possible subranges as well as individual numerical values within that range to the tenth of the unit of the lower limit unless the context clearly dictates otherwise. For example, description of a range such as from 1 to 6 should be considered to have specifically described subranges such as from 1 to 3, from 1 to 4, from 1 to 5, from 2 to 4, from 2 to 6, from 3 to 6 etc., as well as individual values within that range, for example, 1.1, 2, 2.3, 5, and 5.9. This applies regardless of the breadth of the range. The upper and lower limits of these intervening ranges may independently be included in the smaller ranges, and are also encompassed within the invention, subject to any specifically excluded limit in the stated range. Where the stated range includes one or both of the limits, ranges excluding either or both of those included limits are also included in the invention, unless the context clearly dictates otherwise.
  • Unless specifically stated or obvious from context, as used herein, the term “about” in reference to a number or range of numbers is understood to mean the stated number and numbers +/−10% thereof, or 10% below the lower listed limit and 10% above the higher listed limit for the values listed for a range.
  • The terminology used herein is for the purpose of describing particular instances only and is not intended to be limiting of any embodiment. As used herein, the singular forms “a,” “an” and “the” are intended to include the plural forms as well, unless the context clearly indicates otherwise. It will be further understood that the terms “comprises” and/or “comprising,” when used in this specification, specify the presence of stated features, integers, steps, operations, elements, and/or components, but do not preclude the presence or addition of one or more other features, integers, steps, operations, elements, components, and/or groups thereof. As used herein, the term “and/or” includes any and all combinations of one or more of the associated listed items.
  • As used herein, the term “comprising” is inclusive or open-ended and does not exclude additional unrecited elements, device features, compositional components, or method steps.
  • The term “locus” as used herein refers to a discrete region on a structure which provides support for extension of an oligonucleic acid.
  • As used herein, the terms “preselected sequence,” “predefined sequence” or “predetermined sequence” are used interchangeably. The terms refer to sequence of a polymer that is known and chosen before synthesis or assembly of the polymer. In particular, various aspects of the invention are described herein primarily with regard to the preparation of nucleic acid molecules, the sequence of the oligonucleotide or polynucleotide being known and chosen before the synthesis or assembly of the nucleic acid molecules.
  • Substrates, Sub-Fields, Clusters and Loci
  • Provided herein are devices having a substrate (e.g., a plate) for the generation of a library of oligonucleic acids. An exemplary substrate 100 is illustrated in FIG. 1 , wherein the substrate 100 has about the same size dimensions as a standard 96 well plate: 140 mm by 90 mm. The substrate 100 comprises clusters grouped in 24 regions or sub-fields 105, each sub-field 105 comprising an array of 256 clusters 110. An expanded view of an exemplary sub-field 105 is shown in FIG. 2 . In the expanded view of four clusters (FIG. 2 ), a single cluster 110, has a Y axis cluster pitch (distance from center to center of adjacent clusters) of 1079.210 um or 1142.694 um, and an X axis cluster pitch of 1125 um. An illustrative cluster 110 is depicted in FIG. 3 , where the Y axis loci pitch (distance from center to center of adjacent loci) is 63.483 um, and an X axis loci pitch is 75 um. The locus width at the longest part, e.g., diameter for a circular loci, is 50 um and the distance between loci is 24 um. The number of loci 305 in the exemplary cluster in FIG. 3 is 121.
  • Fluid Conditioning
  • Provided herein are substrates comprising features which are segregated to allow for efficient chemical exchange of reagents during de novo oligonucleic acid synthesis. An exemplary arrangement is illustrated in FIGS. 4A-4B where a plate 405 is illustrated comprising a main channel 410 and a plurality of microchannels 415 connected to the main channel 410. The connection between the main channel 410 and the plurality of microchannels 415 provides for a fluid communication for flow paths from the main channel 410 to the each of the plurality of microchannels 415. A plate 405 described herein can comprise multiple main channels 410. The plurality of microchannels 415 collectively forms a cluster within the main channel 410. In some cases, a library of oligonucleic acids is synthesized in a plurality of loci where the loci are collectively a plurality of microchannels 415 of a cluster where the cluster is within a main channel 410, followed by the assembly of the oligonucleic acids into a large nucleic acid such as gene, wherein the assembly of the large nucleic acid optionally occurs within a main channel of the cluster, e.g., by using PCA. In further cases, a different oligonucleic acid is grown in each of the microchannels 415 with a main channel 410, and the oligonucleic acids collectively encode for a single gene.
  • The structure is configured to allow for controlled flow for de novo oligonucleic acid synthesis by providing for rapid exchange of chemical exposure during de novo synthesis of oligonucleic acids. For example, in some cases, configuration described herein provide for the controlled and even distribution of mass transfer paths, chemical exposure times, and/or wash efficiency during oligonucleic acid synthesis. In some instances, the configuration of a substrate allows for increased sweep efficiency, for example by providing sufficient volume for a growing an oligonucleic acid such that the excluded volume by the growing oligonucleic acid does not take up more than 50, 45, 40, 35, 30, 25, 20, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2, 1%, or less of the initially available volume that is available or suitable for growing the oligonucleic acid.
  • In addition to the physical segregation between each of the microchannels 415 of the plurality of microchannels, chemical coatings also provide and additional means for segregating oligonucleic acid species situated within a microchannel. Segregation can be achieved by differential functionalization of the surface, for example by having active and passive regions for oligonucleic acid synthesis coated on the surface. Differential functionalization can also be achieved by alternating the hydrophobicity across the substrate surface, thereby creating water contact angle effects that may cause beading or wetting of the deposited reagents. Employing larger structures can decrease splashing and cross-contamination of distinct oligonucleic acid synthesis locations with reagents of the neighboring spots. A device, such as an oligonucleic acid synthesizer, may be used to deposit reagents to distinct oligonucleic acid synthesis locations.
  • Microchannels: Structural Features
  • Microchannels described herein provide chemical properties, dimensions (width, height/depth, or length) de novo synthesized oligonucleic acids having a low error rate. While the plurality of microchannels 415 in FIG. 4 are circular, various shapes can be used to enhance flow rates through the channel, e.g., microchannels with curves or combs. A microchannel having a shape providing an increased surface area to volume ratio may be desirable for several reasons. First, an increase in surface area provides an increase in area that is suitable for oligonucleic acid attachment and synthesis. Second, a locus in the shape of a microchannel with increased surface area to volume ratio requires less fluid for efficient flow through the channel, thereby allowing less reagent volume per reaction. Third, without wishing to be bound by theory, the efficient flow through a locus in the shape of a microchannel with increased surface area to volume ratio minimizes residual occupation of reagents during flow through the microchannel and enhances wash efficiency, thereby minimizing or essentially eliminating undesirable secondary reactions during the chemical steps involved in the oligonucleotide extension process. Minimizing undesirable secondary reactions is another factor for keeping error rate down during oligonucleic acid synthesis.
  • Exemplary microchannels are illustrated in FIG. 5 which shows a top view of a double comb 510, single comb 520 or serpentine microchannel shape 530. In each exemplary microchannel in FIG. 5 , the width of the microchannel at the narrowest segment is 5 um, and each of the microchannel comprises at least one turn greater than 90 degrees in total. In some cases, the microchannel comprises 1 to 10 or more turns greater than 90 degrees in total. In some cases, the turns are curved and the microchannel comprises 1 to 10 or more turns in total. In the case of the single comb 520 and double comb 510, the turns are 90 degrees, i.e. perpendicular fluid paths from a top view. In the case of the serpentine comb 530, the turns are curved and 180 degrees, i.e. a U turn is viewed from top view. In some cases, a turn in a microchannel fluid path is 45 to 180 degrees in total, when viewed from a top view.
  • In a first example, a main channel 600 comprises a cluster of double comb channels, where each double comb channel 510 has a total width of 57 um, the width of the microchannel at the narrowest segment is 5 um, and each stick of the “comb” is 14 um apart from the center of another stick (FIG. 6 ). In a second example, a main channel 700 comprises a cluster of single comb channels, where each single comb channel 520 has a total width of 49 um, the width of the microchannel at the narrowest segment is 5 um, and each stick of the “comb” is 14 um apart from the center of another stick (FIG. 7 ). In a third example, a main channel 800 comprises a cluster of serpentine shaped channels, where each serpentine shaped channel 530 has a total width of 54 um, the width of the microchannel at the narrowest segment is 5 um, and each turn of the shape results in a parallel region 14 um apart from another region (FIG. 7 ). In some instances, the microchannel extends vertically into the substrate, e.g., a plate.
  • In some instances, a total surface area for a locus (e.g., a microchannel) in a device described herein is greater than about 9000, 10000, 11000, 12000, 12500, 12600, 12700, 12800, 12900 or 13000 um2. In some instances, the total surface area for a locus in a device described herein is about 10000 to about 15000 um2. In some instances, the total surface area for a locus in a device described herein is about 12000 to about 13000 um2. In some instances, the total surface area to volume ratio (1/um) for a locus in a device described herein is about 0.2 to 0.5. In some instances, the total surface area to volume ratio (1/um) for a locus in a device described herein is greater than 0.20. In some instances, the total surface area to volume ratio (1/um) for a locus in a device described herein is greater than about 0.40. In some instances, the total surface area to volume ratio (1/um) for a locus in a device described herein is about 0.40. In some instances, the total surface area to volume ratio (1/um) for a locus in a device described herein is 0.41.
  • In some instances, a microchannel described herein has a width to depth (or height) ratio of 1 to 0.01, wherein the width is a measurement of the width at the narrowest segment of the microchannel. In some instances, a microchannel described herein has a width to depth (or height) ratio of 0.5 to 0.01, wherein the width is a measurement of the width at the narrowest segment of the microchannel. In some instances, a microchannel described herein has a width to depth (or height) ratio of about 0.01, 0.05, 0.1, 0.15, 0.16, 0.2, 0.5, or 1.
  • In some instances, a substrate is provided comprises a plurality of microchannels corresponding to a plurality of loci within a cluster, wherein the height or depth of the microchannel is from about 5 um to about 500 um, from about 5 um to about 400 um, from about 5 um to about 300 um, from about 5 um to about 200 um, from about 5 um to about 100 um, from about 5 um to about 50 um, or from about 10 um to about 50 um. In some cases, the height of a microchannel is less than 100 um, less than 80 um, less than 60 um, less than 40 um or less than 20 um. In some cases, microchannel height is about 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 200, 300, 400, 500 μm or more.
  • In some instances, the width of a locus (e.g., microchannel or microwell) is from about 0.5 μm to about 500 um, from about 1 um to about 200 um, from about 1 um to about 100 um, from about 5 um to about 100 um, or from about 0.1 um to about 100 um, for example, about 90 um, 80 um, 70 um, 60 um, 50 um, 40 um, 30 um, 20 um, 10 um, 5 um, 1 um or 0.5 um. In some instances, the width of a locus (e.g., microchannel) is less than about 100 um, 90 um, 80 um, 70 um, 60 um, 50 um, 40 um, 30 um, 20 um or 10 um. In some instances, the distance between the center of two adjacent loci is from about 1 um to about 500 um, from about 1 um to about 200 um, from about 1 um to about 100 um, from about 5 um to about 200 um, from about 5 um to about 100 um, from about 5 um to about 50 um, or from about 5 um to about 30 um, for example, about 20 um. In some instances, the total width of a microchannel is about 10 um, 20 um, 30 um, 40 um, 50 um, 60 um, 70 um, 80 um, 90 um, or 100 um. In some instances, the total width of a microchannel is about 10 um to 100 um, 30 um to 100 um, or 50 um to 70 um.
  • In some cases, each locus supports the synthesis of a population of oligonucleic acids having a different sequence than a population of oligonucleic acids grown on another locus. Provided herein are surfaces which comprise at least 10, 100, 256, 500, 1000, 2000, 3000, 4000, 5000, 6000, 7000, 8000, 9000, 10000, 11000, 12000, 13000, 14000, 15000, 20000, 30000, 40000, 50000 or more clusters. Provided herein are surfaces which comprise more than 2,000; 5,000; 10,000; 20,000; 30,000; 50,000; 100,000; 200,000; 300,000; 400,000; 500,000; 600,000; 700,000; 800,000; 900,000; 1,000,000; 5,000,000; or 10,000,000 or more distinct loci (e.g., microchannels). In some cases, each cluster includes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 120, 130, 150, 200, 500 or more loci. In some cases, each cluster includes 50 to 500, 50 to 200, 50 to 150, or 100 to 150 loci. In some cases, each cluster includes 100 to 150 loci. In exemplary arrangements, each cluster includes 109, 121, 130 or 137 loci.
  • Provided herein are structures wherein the distance between the centers of two adjacent loci within a cluster is from about 1 um to about 500 um, from about 5 um to about 200 um, or from about 0.5 μm to about 100 um. Provided herein are structures wherein the distance between two centers of adjacent loci is about 0.5 μm, 20 um, 25 um, 30 um, 40 um, 50 um, 60 um, 70 um, 80 um, 90 um, or 100 um.
  • Provided herein are loci having a width at the longest segment of 5 to 100 um. In some cases, the loci have a width at the longest segment of about 30, 35, 40, 45, 50, 55 or 60 um. In some cases, the loci are microchannels having multiple segments, wherein each segment has a center to center distance apart of 5 to 50 um. In some cases, the center to center distance apart for each segment is about 5, 10, 15, 20 or 25 um.
  • Main Channels: Structural Features
  • Main channels described herein extend from a top surface of a substrate, e.g., a plate, into the plate until reaching an interface with a plurality of microchannels, each microchannel connecting to a bottom surface of the substrate. In some instances, the main channel extends vertically. Main channels, e.g., main channel 410, can be in circular, rectangular, tapered, or rounded shapes.
  • In some instances, a width (a diameter in the case of a circle) of a cluster or the width of a main channel comprising a cluster, or both, is between about 0.05 mm to about 50 mm, between about 0.05 mm to about 10 mm, between about 0.05 mm and about 5 mm, between about 0.05 mm and about 4 mm, between about 0.05 mm and about 3 mm, between about 0.05 mm and about 2 mm, between about 0.05 mm and about 1 mm, between about 0.05 mm and about 0.5 mm, between about 0.05 mm and about 0.1 mm, between about 0.1 mm and 10 mm, between about 0.2 mm and 10 mm, between about 0.3 mm and about 10 mm, between about 0.4 mm and about 10 mm, between about 0.5 mm and 10 mm, between about 0.5 mm and about 5 mm, between about 0.5 mm and about 1.5 mm, or between about 0.5 mm and about 2 mm. In some instances, the width of a cluster or main channel or both is less than or about 5 mm, 4 mm, 3 mm, 2 mm, 1.5 mm, 1.2 mm, 1.15 mm, 1 mm, 0.5 mm, 0.1 mm, 0.09 mm, 0.08 mm, 0.07 mm, 0.06 mm or 0.05 mm. In some instances, the width of a cluster or main channel is between about 1.0 and about 1.3 mm. In some instances, the width of a cluster or main channel, or both is about 1.150 mm. In some instances, the width of a cluster or main channel, or both is about 0.08 mm.
  • In some instances, the height (depth) of a main channel is from about 20 um to about 1000 um, from about 50 um to about 1000 um, from about 100 μm to about 1000 um, from about 200 μm to about 1000 um, from about 300 μm to about 1000 um, from about 400 um to about 1000 um, or from about 500 μm to about 1000 um. In some cases, the height of a main channel is less than about 1000 um, less than about 900 um, less than about 800 um, less than about 700 um, or less than about 600 um. In some cases, the height of a main channel is about 500 um. In some cases, the height of a main channel is about 450 um.
  • In some instances, the number of distinct nucleic acids or genes assembled from a plurality of oligonucleic acids synthesized on a substrate is dependent on the number of clusters available in the substrate. In some instances, the density of clusters within a substrate is at least or about 1 cluster per 100 mm2, 1 cluster per 10 mm2, 1 cluster per 5 mm2, 1 cluster per 4 mm2, 1 cluster per 3 mm2, 1 cluster per 2 mm2, 1 cluster per 1 mm2, 2 clusters per 1 mm2, 3 clusters per 1 mm2, 4 clusters per 1 mm2, 5 clusters per 1 mm2, 10 clusters per 1 mm2, 50 clusters per 1 mm2 or more. In some instances, a substrate comprises from about 1 cluster per 10 mm2 to about 10 clusters per 1 mm2. In some instances, the distance between the centers of two adjacent clusters is less than about 50 um, 100 um, 200 um, 500 um, 1000 um, or 2000 um or 5000 um. In some cases, the distance between the centers of two adjacent clusters is between about 50 um and about 100 um, between about 50 um and about 200 um, between about 50 um and about 300 um, between about 50 um and about 500 um, and between about 100 μm to about 2000 um. In some cases, the distance between the centers of two adjacent clusters is between about 0.05 mm to about 50 mm, between about 0.05 mm to about 10 mm, between about 0.05 mm and about 5 mm, between about 0.05 mm and about 4 mm, between about 0.05 mm and about 3 mm, between about 0.05 mm and about 2 mm, between about 0.1 mm and 10 mm, between about 0.2 mm and 10 mm, between about 0.3 mm and about 10 mm, between about 0.4 mm and about 10 mm, between about 0.5 mm and 10 mm, between about 0.5 mm and about 5 mm, or between about 0.5 mm and about 2 mm.
  • In some instances, the number of distinct oligonucleic acids synthesized on a substrate is dependent on the number of distinct loci available in the substrate. In some instances, the density of loci within a cluster of a substrate is at least or about 1 locus per mm2, 10 loci per mm2, 25 loci per mm2, 50 loci per mm2, 65 loci per mm2, 75 loci per mm2, 100 loci per mm2, 130 loci per mm2, 150 loci per mm2, 175 loci per mm2, 200 loci per mm2, 300 loci per mm2, 400 loci per mm2, 500 loci per mm2, 1,000 loci per mm2 or more. In some cases, a substrate comprises from about 10 loci per mm2 to about 500 mm2, from about 25 loci per mm2 to about 400 mm2, from about 50 loci per mm2 to about 500 mm2, from about 100 loci per mm2 to about 500 mm2, from about 150 loci per mm2 to about 500 mm2, from about 10 loci per mm2 to about 250 mm2, from about 50 loci per mm2 to about 250 mm2, from about 10 loci per mm2 to about 200 mm2, or from about 50 loci per mm2 to about 200 mm2. In some instances, the distance between the centers of two adjacent loci within a cluster is from about 10 um to about 500 um, from about 10 um to about 200 um, or from about 10 um to about 100 um. In some cases, the distance between two centers of adjacent loci is greater than about 10 um, 20 um, 30 um, 40 um, 50 um, 60 um, 70 um, 80 um, 90 um or 100 um. In some cases, the distance between the centers of two adjacent loci is less than about 200 um, 150 um, 100 um, 80 um, 70 um, 60 um, 50 um, 40 um, 30 um, 20 um or 10 um.
  • A device described herein may comprise multiple main channels. In some cases, a device described herein comprises 1 to 250, 2 to 250, 1 to 500 or more main channels. In some cases, a device described herein comprises about 2, 10, 50, 100, 150, 200, 250, 256, 500, 512, 1000, 2500, 3000, 4000, 5000, 6000, 6144, 10000 or more main channels. In some cases, a plate described herein comprises about 2, 10, 50, 100, 150, 200, 250, 256, 500, 512, 1000, 2500, 3000, 4000, 5000, 6000, 6144, 10000 or more main channels. In some cases, the plate is a silicon plate or a silicon on insulator (SOI) plate.
  • In some instances, a substrate comprises a surface that supports the synthesis of a plurality of oligonucleic acids having different predetermined sequences at addressable locations on a common support. In some instances, a substrate described herein provides support for the synthesis of more than 2,000; 5,000; 10,000; 20,000; 30,000; 50,000; 100,000; 200,000; 300,000; 400,000; 500,000; 600,000; 700,000; 800,000; 900,000; 1,000,000; 1,200,000; 1,400,000; 1,600,000; 1,800,000; 2,000,000; 2,500,000; 3,000,000; 3,500,000; 4,000,000; 4,500,000; 5,000,000; 10,000,000 or more non-identical oligonucleic acids. In some cases, the substrate provides support for the synthesis of more than 2,000; 5,000; 10,000; 20,000; 50,000; 100,000; 200,000; 300,000; 400,000; 500,000; 600,000; 700,000; 800,000; 900,000; 1,000,000; 1,200,000; 1,400,000; 1,600,000; 1,800,000; 2,000,000; 2,500,000; 3,000,000; 3,500,000; 4,000,000; 4,500,000; 5,000,000; 10,000,000 or more oligonucleic acids encoding for distinct sequences. In some instances, at least a portion of the oligonucleic acids have an identical sequence or are configured to be synthesized with an identical sequence. In some instances, the substrate provides a surface environment for the growth of oligonucleic acids having at least about 50, 60, 70, 75, 80, 85, 90, 95, 100, 110, 120, 130, 140, 150, 160, 175, 200, 225, 250, 275, 300, 325, 350, 375, 400, 425, 450, 475, 500 bases or more.
  • In some instances, oligonucleic acids are synthesized on distinct loci of a substrate, wherein each locus supports the synthesis of a population of oligonucleic acids. In some cases, each locus supports the synthesis of a population of oligonucleic acids having a different sequence than a population of oligonucleic acids grown on another locus. In some instances, the loci of a substrate are located within a plurality of clusters. In some instances, a substrate comprises at least 10, 500, 1000, 2000, 3000, 4000, 5000, 6000, 7000, 8000, 9000, 10000, 11000, 12000, 13000, 14000, 15000, 20000, 30000, 40000, 50000 or more clusters. In some instances, a substrate comprises more than 2,000; 5,000; 10,000; 100,000; 200,000; 300,000; 400,000; 500,000; 600,000; 700,000; 800,000; 900,000; 1,000,000; 1,100,000; 1,200,000; 1,300,000; 1,400,000; 1,500,000; 1,600,000; 1,700,000; 1,800,000; 1,900,000; 2,000,000; 300,000; 400,000; 500,000; 600,000; 700,000; 800,000; 900,000; 1,000,000; 1,200,000; 1,400,000; 1,600,000; 1,800,000; 2,000,000; 2,500,000; 3,000,000; 3,500,000; 4,000,000; 4,500,000; 5,000,000; or 10,000,000 or more distinct loci. In some cases, each cluster includes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 120, 130, 150 or more loci. In some instances, each cluster includes 50 to 500, 100 to 150, or 100 to 200 loci. In some instances, each cluster includes 109, 121, 130 or 137 loci. In some instances, each cluster includes 5, 6, 7, 8, 9, 10, 11 or 12 loci.
  • In some instances, oligonucleic acids from distinct loci within one cluster have sequences that, when assembled, encode for a contiguous longer oligonucleic acid of a predetermined sequence, for example, a gene. In some cases, a substrate comprising more than 20,000 loci (e.g., microchannels) is used for the synthesis of up to 200 distinct genes of predetermined sequence. In some cases, a substrate comprising more than 29,000 loci (e.g., microchannels) is used for the synthesis of about 240 distinct genes of predetermined sequence. In some cases, a substrate comprising more than 700,000 loci is used for the synthesis of about 6,000 distinct genes of predetermined sequence.
  • Substrate: Materials
  • Substrates provided may be fabricated from a variety of materials suitable for the methods and compositions described herein. In certain instances, substrate materials are fabricated to exhibit a low level of nucleotide binding. In some cases, substrate materials are modified to generate distinct surfaces that exhibit a high level of nucleotide binding. In some instances, substrate materials are transparent to visible and/or UV light. In some instances, substrate materials are sufficiently conductive, e.g., are able to form uniform electric fields across all or a portion of a substrate. In some instances, conductive materials may be connected to an electric ground. In some cases, the substrate is heat conductive or insulated. In some cases, the materials are chemical resistant and heat resistant to support chemical or biochemical reactions, for example oligonucleic acid synthesis reaction processes. In some instances, a substrate comprises flexible materials. Flexible materials include, without limitation, modified nylon, unmodified nylon, nitrocellulose, polypropylene, and the like. In some instances, a substrate comprises rigid materials. Rigid materials include, without limitation, glass, fuse silica, silicon, silicon dioxide, silicon nitride, plastics (for example, polytetraflouroethylene, polypropylene, polystyrene, polycarbonate, and blends thereof, and the like), and metals (for example, gold, platinum, and the like). In some instances, a substrate is fabricated from a material comprising silicon, polystyrene, agarose, dextran, cellulosic polymers, polyacrylamides, polydimethylsiloxane (PDMS), glass, or any combination thereof. The substrates may be manufactured with a combination of materials listed herein or any other suitable material known in the art. In some instances, substrates described herein are in the shape of a plate, film or tape.
  • Substrate: Structural Features
  • In some instances, a substrate is about the size of a standard 96 well plate, for example between about 100 and 200 mm by between about 50 and 150 mm. In some instances, a substrate has a diameter less than or equal to about 1000 mm, 500 mm, 450 mm, 400 mm, 300 mm, 250 nm, 200 mm, 150 mm, 100 mm or 50 mm. In some instances, the diameter of a substrate is between about 25 mm and 1000 mm, between about 25 mm and about 800 mm, between about 25 mm and about 600 mm, between about 25 mm and about 500 mm, between about 25 mm and about 400 mm, between about 25 mm and about 300 mm, or between about 25 mm and about 200. Non-limiting examples of substrate size include about 300 mm, 200 mm, 150 mm, 130 mm, 100 mm, 76 mm, 51 mm and 25 mm. In some instances, a substrate has a planar surface area of at least about 100 mm2; 200 mm2; 500 mm2; 1,000 mm2; 2,000 mm2; 5,000 mm2; 10,000 mm2; 12,000 mm2; 15,000 mm2; 20,000 mm2; 30,000 mm2; 40,000 mm2; 50,000 mm2 or more. In some instances, the thickness of a substrate is between about 50 mm and about 2000 mm, between about 50 mm and about 1000 mm, between about 100 mm and about 1000 mm, between about 200 mm and about 1000 mm, or between about 250 mm and about 1000 mm. Non-limiting examples of substrate thickness include 275 mm, 375 mm, 525 mm, 625 mm, 675 mm, 725 mm, 775 mm and 925 mm. In some cases, the thickness of a substrate varies with diameter and depends on the composition of the substrate. For example, a substrate comprising materials other than silicon may have a different thickness than a silicon substrate of the same diameter. Substrate thickness may be determined by the mechanical strength of the material used and the substrate must be thick enough to support its own weight without cracking during handling.
  • In some instances, a substrate described herein comprises a plurality of smaller regions, for example, at least about 2, 4, 6, 8, 10, 16, 24, 39, 50, 100 or more regions, wherein each region may be used independently from another region. In some cases, regions of a substrate are sub-fields or chips of a substrate. In some instances, reference to a substrate includes a region of a substrate.
  • Active and Passive Functionalization
  • Selective deposition or selective functionalization refers to a process that produces two or more distinct areas on a structure, wherein at least one area has a different surface or chemical property that another area of the same structure. Such properties include, without limitation, surface energy, chemical termination, surface concentration of a chemical moiety, and the like. In some cases, active functionalization refers to a method comprising the functionalization of a surface that will be utilized for oligonucleic acid synthesis. In some cases, passive functionalization refers to a method comprising the functionalization of a surface that will render these areas ineffective for oligonucleic acid synthesis. Any suitable process that changes the chemical properties of the surface described herein or known in the art may be used to functionalize the surface, for example chemical vapor deposition of an organosilane. Typically, this results in the deposition of a self-assembled monolayer (SAM) of the functionalization species.
  • In some instances, a method for functionalizing a surface of a substrate for oligonucleic acid synthesis comprises a resist or photoresist coat. Photoresist, in many cases, refers to a light-sensitive material useful in photolithography to form patterned coatings. It can be applied as a liquid to solidify on a substrate as volatile solvents in the mixture evaporate. In some instances, the resist is applied in a spin coating process as a thin film, e.g., 1 um to 100 um. In some cases, the coated resist is patterned by exposing it to light through a mask or reticle, changing its dissolution rate in a developer. In some cases, the resist cost is used as a sacrificial layer that serves as a blocking layer for subsequent steps that modify the underlying surface, e.g., etching, and then is removed by resist stripping. In some instances, the flow of resist throughout various features of the structure is controlled by the design of the structure. In some instances, a surface of a structure is functionalized while areas covered in resist are protected from active or passive functionalization.
  • In some instances, a substrate suitable for functionalization (e.g., an etched substrate comprising three-dimensional features) is deposited with resist, for example, by an oligonucleic acid synthesizer devices capable of delivering drops of fluid with micrometer accuracy. To resist coat only a small region of the substrate (e.g., lowered features such as a well and/or channel), a droplet of resist may be deposited into the lowered feature where it optionally spreads. In some instances, a portion of the resist is removed, for example, by etching (e.g., oxygen plasma etch) to leave a smooth surface covering only a select area. In some instances, the substrate is passively functionalized with a passive functionalization agent comprising a chemically inert moiety to create a surface having low surface energy. In some instances, the passively functionalized substrate is resist stripped, exposing areas of the substrate that were exposed during front-end processing (etched regions).
  • Described herein are methods for the synthesis of oligonucleic acids having a low error rate compared to a predetermined sequence using a device configured to regulate the flow of reagents in a microfluidic environment. An exemplary method is illustrated in FIGS. 9A-9F, which depicts a workflow for active functionalization of a microchannels and passive functionalization of surrounding areas, e.g., main channels. An etched substrate 905 is prepared for active functionalization. In FIG. 9A, the substrate is wet cleaned, for example, using a piranha solution. In some instances, the substrate is plasma cleaned, for example, by dry oxygen plasma exposure. In FIG. 9B, the device layer is coated with photoresist 910, optionally after cleaning. In some instances, the photoresist is coated by a process governed by wicking into the device layer channels. In some instances, the photoresist is patterned using photolithography to expose areas that are desired to be passive (i.e., areas where oligonucleic acid synthesis is not designed to take place). Patterning by photolithography may occur by exposing the resist to light through a binary mask that has a pattern of interest. After exposure, the resist in the exposed regions may be removed in developer solution in FIG. 9C, and leaves photoresist at predetermined regions 915. A subsequent step, as shown in FIG. 9D, involves exposure to a passive functionalization agent 920 such as a low surface energy silane (e.g., fluorosilane gas vapor), for example, by chemical vapor deposition (CVD). Exposure to fluorosilane gas results in the deposition of a fluorocarbon on the surfaces without photoresist. In some cases, the substrate is exposed to a hydrocarbon silane. In some instances, passive functionalization comprises exposing a substrate to a passive functionalization agent such as one comprising silane. In some instances, the passively functionalized substrate are unresponsive to additional layers of functionalization agent (e.g., active functionalization agent) creating a monolayer on the surface. A substrate that has been coated with resist, and optionally patterned by photolithography and/or optionally passively functionalized, in many cases, is resist stripped. Resist stripping as shown in FIG. 9E leaves some surfaces passively functionalized while exposing regions of the substrate that was underneath the resist 925. For example, a resist is dissolved in an organic solvent. As another example, resist stripping of a substrate that was passively functionalized with a fluorosilane gas, leaves a surface having some regions of fluorination. In some cases, regions that were underneath the resist comprise silicon or silicon dioxide. In FIG. 9F, the surface is actively functionalized to prepare the surface for oligonucleic acid synthesis. An exemplary active functionalization agent is one that has a higher surface energy than the passive functionalization agent.
  • An exemplary workflow for the generation of differential functionalization patterns of a substrate is described herein, FIGS. 9A-F. The following workflow is an example process and any step or component may be omitted or changed in accordance with properties desired of the final functionalized substrate. In some cases, additional components and/or process steps are added to the process workflows embodied herein. In some instances, a substrate is first cleaned, for example, using a piranha solution. An example of a cleaning process includes soaking a substrate in a piranha solution (e.g., 90% H2SO4, 10% H2O2) at an elevated temperature (e.g., 120° C.) and washing (e.g., water) and drying the substrate (e.g., nitrogen gas). The process optionally includes a post piranha treatment comprising soaking the piranha treated substrate in a basic solution (e.g., NH4OH) followed by an aqueous wash (e.g., water). In some instances, a substrate is plasma cleaned, optionally following the piranha soak and optional post piranha treatment. An example of a plasma cleaning process comprises an oxygen plasma etch. In some instances, the surface is deposited with an active functionalization agent following by vaporization. In some instances, the substrate is actively functionalized prior to cleaning, for example, by piranha treatment and/or plasma cleaning.
  • The process for substrate functionalization optionally comprises a resist coat and a resist strip. In some instances, following active surface functionalization, the substrate is spin coated with a resist, for example, SPR™ 3612 positive photoresist. The process for substrate functionalization, in various instances, comprises lithography with patterned functionalization. In some instances, photolithography is performed following resist coating. In some instances, after lithography, the substrate is visually inspected for lithography defects. The process for substrate functionalization, in some instances, comprises a cleaning step, whereby residues of the substrate are removed, for example, by plasma cleaning or etching. In some instances, the plasma cleaning step is performed at some step after the lithography step.
  • In some instances, a substrate coated with a resist is treated to remove the resist, for example, after functionalization and/or after lithography. In some cases, the resist is removed with a solvent, for example, with a stripping solution comprising N-methyl-2-pyrrolidone. In some cases, resist stripping comprises sonication or ultrasonication. In some instances, a resist is coated and stripped, followed by active functionalization of the exposed areas to create a desired differential functionalization pattern.
  • In various instances, the methods and compositions described herein relate to the application of photoresist for the generation of modified surface properties in selective areas, wherein the application of the photoresist relies on the fluidic properties of the substrates defining the spatial distribution of the photoresist. Without being bound by theory, surface tension effects related to the applied fluid may define the flow of the photoresist. For example, surface tension and/or capillary action effects may facilitate drawing of the photoresist into small structures in a controlled fashion before the resist solvents evaporate. In some instances, resist contact points are pinned by sharp edges, thereby controlling the advance of the fluid. The underlying structures may be designed based on the desired flow patterns that are used to apply photoresist during the manufacturing and functionalization processes. A solid organic layer left behind after solvents evaporate may be used to pursue the subsequent steps of the manufacturing process. Substrates may be designed to control the flow of fluids by facilitating or inhibiting wicking effects into neighboring fluidic paths. For example, a substrate is designed to avoid overlap between top and bottom edges, which facilitates the keeping of the fluid in top structures allowing for a particular disposition of the resist. In an alternative example, the top and bottom edges overlap, leading to the wicking of the applied fluid into bottom structures. Appropriate designs may be selected accordingly, depending on the desired application of the resist.
  • As illustrated in the detailed cross view of FIG. 10A, an exemplary described herein is coated with a layer of material comprising on or more active functionalization agent. FIG. 10A illustrates the deposition of reagents 1005 on a plate 405. The substrate 405 comprises a plurality of microchannels 415 in fluidic connection with a main channel 410. In this exemplary device, a single main channel 410 is depicted, and the dashed lines indicate that this is just one of many main channel 410/plurality of microchannels 415 connections in a single plate 405. Each of the plurality of microchannels 415 is coated with an active functionalization agent 1015. Each microchannel 415 of the plurality of microchannels in the example has a width of 5 um and a depth of 30 um. The main channel 410 has a diameter of 1150 um. The total height of the plate is 450 um. The main channel 410 encircles a cluster of microchannels. In some instances, as illustrated in FIG. 10B, a substrate described herein is coated with a layer of material comprising on or more passive functionalization agents. In this exemplary device, a passive functionalization agent 1020 coats surface of a plate 405. In some instances, as illustrated in FIG. 10C, a substrate described herein is coated with a layer of material comprising on or more passive functionalization agents and the microchannels of the substrate are coated with one or more active functionalization agents. In another exemplary device, the starting substrate is a silicon on insulator plate, where layers of silicon sandwich an insulator layer 1110, typically silicon dioxide. In some instances, the thickness of the insulator layer 1110 is from 1 to 50 um, e.g., 20 um. The remaining feature of this exemplary device, as shown in FIGS. 11A-11C, as the same as those depicted in FIGS. 10A-10C.
  • In some cases, only loci (i.e., microchannels) in a device described herein are coated with active functionalization agent. An active functionalization agent is a molecule that binds to the surface of the substrate and is also capable of binding to a nucleic acid monomer, thereby supporting a coupling reaction to the surface. Exemplary active functionalization agents are molecules having a hydroxyl group available for coupling with a nucleoside in a coupling reaction. In some cases, only main channels and/or surrounding areas (and not the microchannels) in a device described herein are coated with passive functionalization agent. A passive functionalization agent is a molecule that binds to the surface of the substrate and lacks a moiety available for coupling with a nucleoside in a coupling reaction.
  • Oligonucleic acids synthesized in the channels may be released for the purposes of generating longer nucleic acids. In some cases, following oligonucleic acid synthesis, oligonucleic acids within one cluster are released from their respective surfaces and pool into the main channel. In some cases, the pooled oligonucleic acids are assembled into a larger nucleic acid, such as a gene, within the main channel, so that the main channel functions as a reactor for nucleic acid assembly. In other cases, following oligonucleic acid synthesis, oligonucleic acids within one cluster are released from their respective surfaces and pool into a nanoreactor in fluidic communication with the microchannels.
  • In some embodiments, nucleic acid verification (e.g., sequencing of oligonucleic acids and/or assembled genes) is performed within a reactor or well. Nucleic acid assembly includes polymerase cycling assembly (PCA). In some cases, a capping element or other device is placed over open sides of the main channel to create an enclosed reactor. A substrate comprising a main channel that functions as a reactor for each cluster has the advantage that each cluster may have a different environment from another cluster in another reactor. As an example, sealed reactors (e.g., those with capping elements) may experience controlled humidity, pressure or gas content.
  • In some instances, a substrate is configured for both active and passive functionalization moieties bound to the surface at different areas of the substrate surface, generating distinct regions for oligonucleic acid synthesis to take place. In some instances, both active and passive functionalization agents are mixed within a particular region of the surface. Such a mixture provides a diluted region of active functionalization agent and therefore lowers the density of functionalization agent in a particular region.
  • Substrates described herein may comprise a high surface energy region at the site of active functionalization agent deposition. In some instances, the high surface energy region is coated with aminosilane. The silane group binds to the surface, while the rest of the molecule provides a distance from the surface and a free group at the end to which incoming bases attach. In some instances, the free group is a hydroxyl group. In some instances the high surface energy region includes an active functionalization reagent, e.g., a chemical that binds the substrate efficiently and also couples efficiently to monomeric nucleic acid molecules. In some cases, such molecules have a hydroxyl group which is available for interacting with a nucleoside in a coupling reaction. In some instances, the amino silane is selected from the group consisting of 11-acetoxyundecyltriethoxysilane, n-decyltriethoxysilane, (3-aminopropyl)trimethoxysilane, (3-aminopropyl)triethoxysilane, glycidyloxypropyl/trimethoxysilane and N-(3-triethoxysilylpropyl)-4-hydroxybutyramide. In some instances the high surface energy region includes a passive functionalization reagent, e.g., a chemical that binds the substrate efficiently but does not couple efficiently to monomeric nucleic acid molecules.
  • In some instances, described herein are substrates comprising a plurality of clusters, wherein each cluster comprises a plurality of loci that support the attachment and synthesis of oligonucleic acids. In some instances, a locus is on a three-dimensional surface, e.g., a well, microchannel, channel, or post. In some instances, a surface of a locus comprises a material that is actively functionalized to attach to at least one nucleotide for oligonucleic acid synthesis, or preferably, a population of identical nucleotides for synthesis of a population of oligonucleic acids. In some instances, oligonucleic acid refers to a population of oligonucleic acids encoding for the same nucleic acid sequence. In some cases, a surface of a substrate is inclusive of one or a plurality of surfaces of a substrate.
  • In some cases, the addition of a chemical layer on top of a surface (referred to as adhesion promoter) facilitates structured patterning of loci on a surface of a substrate. Exemplary surfaces which can benefit from adhesion promotion include, without limitation, glass, silicon, silicon dioxide and silicon nitride. In some cases, the adhesion promoter is a chemical with a high surface energy. In some instances, a second chemical layer is deposited on a surface of a substrate. In some cases, the second chemical layer has a low surface energy. The surface energy of a chemical layer coated on a surface can facilitate localization of droplets on the surface. Depending on the patterning arrangement selected, the proximity of loci and/or area of fluid contact at the loci can be altered.
  • In some instances, a substrate surface, or resolved loci, onto which nucleic acids or other moieties are deposited, e.g., for oligonucleic acid synthesis, are smooth or substantially planar or have raised or lowered features. In some instances, a substrate surface is modified with one or more different layers of compounds. Such modification layers of interest include, without limitation, inorganic and organic layers such as metals, metal oxides, polymers, small organic molecules and the like. Non-limiting polymeric layers include peptides, proteins, nucleic acids or mimetics thereof (e.g., peptide nucleic acids and the like), polysaccharides, phospholipids, polyurethanes, polyesters, polycarbonates, polyureas, polyamides, polyetheyleneamines, polyarylene sulfides, polysiloxanes, polyimides, polyacetates, and any other suitable compounds described herein or otherwise known in the art. In some cases, polymers are heteropolymeric. In some cases, polymers are homopolymeric. In some cases, polymers comprise functional moieties or are conjugated.
  • In some instances, resolved loci of a substrate are functionalized with one or more moieties that increase and/or decrease surface energy. In some cases, a moiety is chemically inert. In some cases, a moiety is configured to support a desired chemical reaction, for example, one or more processes in an oligonucleic acid synthesis reaction. The surface energy, or hydrophobicity, of a surface is a factor for determining the affinity of a nucleotide to attach onto the surface. In some instances, a method for substrate functionalization comprises: (a) providing a substrate (e.g., a plate); and (b) silanizing the loci (e.g., microchannels) with a suitable silanizing agent described herein or otherwise known in the art, for example, an organofunctional alkoxysilane molecule. In some cases, the organofunctional alkoxysilane molecule comprises dimethylchloro-octodecyl-silane, methyldichloro-octodecyl-silane, trichloro-octodecyl-silane, trimethyl-octodecyl-silane, triethyl-octodecyl-silane, or any combination thereof. In some instances, a substrate surface comprises functionalized with polyethylene/polypropylene (functionalized by gamma irradiation or chromic acid oxidation, and reduction to hydroxyalkyl surface), highly crosslinked polystyrene-divinylbenzene (derivatized by chloromethylation, and aminated to benzylamine functional surface), nylon (the terminal aminohexyl groups are directly reactive), or etched with reduced polytetrafluoroethylene.
  • In some instances, a substrate surface is functionalized by contact with a derivatizing composition that contains a mixture of silanes, under reaction conditions effective to couple the silanes to the substrate surface, typically via reactive hydrophilic moieties present on the substrate surface. Silanization generally can be used to cover a surface through self-assembly with organofunctional alkoxysilane molecules. A variety of siloxane functionalizing reagents can further be used as currently known in the art, e.g., for lowering or increasing surface energy. The organofunctional alkoxysilanes are classified according to their organic functions. Non-limiting examples of siloxane functionalizing reagents include hydroxyalkyl siloxanes (silylate surface, functionalizing with diborane and oxidizing the alcohol by hydrogen peroxide), diol (dihydroxyalkyl) siloxanes (silylate surface, and hydrolyzing to diol), aminoalkyl siloxanes (amines require no intermediate functionalizing step), glycidoxysilanes (3-glycidoxypropyl-dimethyl-ethoxysilane, glycidoxy-trimethoxysilane), mercaptosilanes (3-mercaptopropyl-trimethoxysilane, 3-4 epoxycyclohexyl-ethyltrimethoxysilane or 3-mercaptopropyl-methyl-dimethoxysilane), bicyclohepthenyl-trichlorosilane, butyl-aldehydr-trimethoxysilane, or dimeric secondary aminoalkyl siloxanes. The hydroxyalkyl siloxanes can include allyl trichlorochlorosilane turning into 3-hydroxypropyl, or 7-oct-1-enyl trichlorochlorosilane turning into 8-hydroxyoctyl. The diol (dihydroxyalkyl) siloxanes include glycidyl trimethoxysilane-derived (2,3-dihydroxypropyloxy)propyl (GOPS). The aminoalkyl siloxanes include 3-aminopropyl trimethoxysilane turning into 3-aminopropyl (3-aminopropyl-triethoxysilane, 3-aminopropyl-diethoxy-methylsilane, 3-aminopropyl-dimethyl-ethoxysilane, or 3-aminopropyl-trimethoxysilane). The dimeric secondary aminoalkyl siloxanes can be bis (3-trimethoxysilylpropyl) amine turning into bis(silyloxylpropyl)amine. In some instances, the functionalizing agent comprises 11-acetoxyundecyltriethoxysilane, n-decyltriethoxysilane, (3-aminopropyl)trimethoxysilane, (3-aminopropyl)triethoxysilane, glycidyloxypropyl/trimethoxysilane and N-(3-triethoxysilylpropyl)-4-hydroxybutyramide.
  • In some instances, a substrate surface is contacting with a mixture of functionalization groups, e.g., aminosilanes, which can be in any different ratio. In some instances, a mixture comprises at least 2, 3, 4, 5 or more different types of functionalization agents. In some instances, the mixture comprises 1, 2, 3 or more silanes. In cases, the ratio of the at least two types of surface functionalization agents in a mixture is about 1:1, 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, 1:10, 2:3, 2:5, 2:7, 2:9, 2:11, 2:13, 2:15, 2:17, 2:19, 3:5, 3:7, 3:8, 3:10, 3:11, 3:13, 3:14, 3:16, 3:17, 3:19, 4:5, 4:7, 4:9, 4:11, 4:13, 4:15, 4:17, 4:19, 5:6, 5:8, 5:9, 5:11, 5:12, 5:13, 5:14, 5:16, 5:17, 5:18, 5:19, 6:7, 6:11, 6:13, 6:17, 6:19, 7:8, 7:9, 7:10, 7:11, 7:12, 7:13, 7:15, 7:16, 7:18, 7:19, 8:9, 8:11, 8:13, 8:15, 8:17, 8:19, 9:10, 9:11, 9:13, 9:14, 9:16, 9:17, 9:19, 10:11, 10:13, 10:17, 10:19, 11:12, 11:13, 11:14, 11:15, 11:16, 11:17, 11:18, 11:19, 11:20, 12:13, 12:17, 12:19, 13:14, 13:15, 13:16, 13:17, 13:18, 13:19, 13:20, 14:15, 14:17, 14:19, 15:16, 15:17, 15:19, 16:17, 16:19, 17:18, 17:19, 17:20, 18:19, 19:20, or any other ratio to achieve a desired surface representation of two groups. In some instances, a ratio of silanes is about 1:100, 1:1000, 1:2000 or 1:3000.
  • In some cases, an active functionalization agent comprises 11-acetoxyundecyltriethoxysilane. In some cases, an active functionalization agent comprises n-decyltriethoxysilane. In some cases, the active functionalization areas comprise one or more different species of silanes, for example, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more silanes. In some cases, one of the one or more silanes is present in the functionalization composition in an amount greater than another silane. For example, a mixed silane solution having two silanes comprises a 99:1, 98:2, 97:3, 96:4, 95:5, 94:6, 93:7, 92:8, 91:9, 90:10, 89:11, 88:12, 87:13, 86:14, 85:15, 84:16, 83:17, 82:18, 81:19, 80:20, 75:25, 70:30, 65:35, 60:40, 55:45 ratio of one silane to another silane. In some instances, an active functionalization agent comprises 11-acetoxyundecyltriethoxysilane and n-decyltriethoxysilane (e.g., in a ratio from about 20:80 to about 1:99, or about 10:90 to about 2:98, or preferably about 5:95). In some cases, an active functionalization agent comprises glycidyloxypropyltriethoxysilane (GOPS). In some instances, the silane is a fluorosilane. In some instances, the silane is a hydrocarbon silane. In some cases, the silane is 3-iodo-propyltrimethoxysilane. In some cases, the silane is octylchlorosilane. In some instances, an active functionalization agent comprises N-(3-triethosysilylpropyl)-4-hydroxybutyramide. In some cases, the passive functionalization agent comprises a silane. In some cases, the passive functionalization agent comprises a mixture of silanes. In some cases, the passive functionalization agent comprises perfluorooctyltrichlorosilane.
  • In some instances, desired surface tensions, wettabilities, water contact angles, and/or contact angles for other suitable solvents are achieved by providing a substrate surface with a suitable ratio of functionalization agents. In some cases, the agents in a mixture are chosen from suitable reactive and inert moieties, thus diluting the surface density of reactive groups to a desired level for downstream reactions. In some instances, the density of the fraction of a surface functional group that reacts to form a growing oligonucleotide in an oligonucleotide synthesis reaction is about 0.005, 0.01, 0.05, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1.0, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2.0, 2.1, 2.2, 2.3, 2.4, 2.5, 3.0, 3.5, 4.0, 4.5, 5.0, 7.0, 10.0, 15.0, 20.0, 50.0, 75.0, 100.0 μMol/m2.
  • In some instances, a surface of a substrate is prepared to have a low surface energy. In some cases, a region of a surface of a substrate described herein is functionalized to enable covalent binding of molecular moieties that can lower the surface energy so that wettability can be reduced. In some instances, a region of surface of a substrate described herein is prepared to have a high surface energy and increased wettability. In some instances, a surface is modified to have a higher surface energy, or become more hydrophilic with a coating where the coating includes molecules having reactive hydrophilic moieties. By altering the surface energy of different parts of a substrate surface, spreading of a deposited reagent liquid (e.g., a reagent deposited during an oligonucleic acid synthesis method) can be adjusted, in some cases facilitated. In some instances, a droplet of reagent is deposited over a predetermined area of a surface with high surface energy. The liquid droplet can spread over and fill a small surface area having a higher surface energy as compared to a nearby surface. In some instances, a substrate surface is modified to comprise reactive hydrophilic moieties such as hydroxyl groups, carboxyl groups, thiol groups, and/or substituted or unsubstituted amino groups. Suitable materials include, but are not limited to, supports that can be used for solid phase chemical synthesis, e.g., cross-linked polymeric materials (e.g., divinylbenzene styrene-based polymers), agarose (e.g., Sepharose®), dextran (e.g., Sephadex®), cellulosic polymers, polyacrylamides, silica, glass (particularly controlled pore glass, or “CPG”), ceramics, and the like. The supports may be obtained commercially and used as is, or they may be treated or coated prior to functionalization.
  • In some instances, provided herein are methods for the manufacture of a substrate using a multilayer activation process. In some instances, one or more layers deposited during a multilayer activation process comprise one or more silanes. In some instances, a substrate, e.g., a silicon plate, is treated with a first layer of a material that modifies the surface to allow for adhesion of a photoresist. Non-limiting examples of materials for surface modification include 11-acetoxyundecyltriethoxysilane, n-decyltriethoxysilane, (3-aminopropyl)trimethoxysilane, (3-aminopropyl)triethoxysilane, glycidyloxypropyl/trimethoxysilane and N-(3-triethoxysilylpropyl)-4-hydroxybutyramide, and mixtures thereof, and/or other suitable materials described elsewhere herein or known in the art. In some instances, the modified substrate is treated with resist, exposed, developed, and residual resist is removed by plasma cleaning, exemplary details of which are described previously herein. In some instances, the substrate is passively functionalized, for example, with fluorosilane, to generate hydrophobic regions. In some instances, the substrate is stripped to remove remaining resist. In some instances, the first modified layer of the substrate is activated to change one or more functional groups of the surface modification material to a hydroxyl group. In a subsequent step, the hydroxyl surface of the substrate is treated with a second layer of surface modification material to dilute surface functional groups and increase coupling efficiency for nucleotide attachment. In some instances, the second layer is activated to change one or more functional groups to a hydroxyl group to support oligonucleic acid synthesis. In some instances, steps comprising the addition of a surface modification material followed by subsequent activation are repeated for one or more additional cycles (e.g., 1, 2, 3, 4, 5 or more additional cycles) to provide optimal spacing between oligonucleic acids during synthesis. In some instances, second and subsequent layers have purely organic chemistries of the general form A-R1-B and C—R2-D where A reacts with the terminal OH group. The system is then purged and C—R2-D reacts with the B group. R1 and R2 chemistries can be repeated to yield the desired film thickness in a molecular layer deposition process. The terminal groups B and D can be hydroxyl or could be converted to hydroxyl in the final step of the deposition.
  • In some cases, the substrate may be two-dimensional (e.g., substantially planar) or three-dimension (e.g., comprise wells and/or channels). In some instances, an actively functionalized surface comprises a specific concentration of hydroxyl groups to achieve a pre-determined surface density for oligonucleic acid synthesis. In some instances, active functionalization is achieved by a wet process using a solution comprising an active functionalization agent. In some cases, the active functionalization agent comprises a silane or mixed silanes. In an example, a surface to be actively functionalized is treated with a solution comprising an active functionalization agent (e.g., 1% solution of N-(3-triethoxysilylpropyl-4hydroxybutyramide in ethanol and acetic acid) and the substrate incubated at a high temperature (e.g., 150° C. for 14 hours). In another example, a chemical vapor deposition process is employed wherein an active functionalization agent is delivered to the surface in a gaseous state. In some cases, an active functionalization agent is delivered by CVD with a controlled deposition pressure (e.g., 200 mTor) and temperature (e.g., 150° C.). A CVD process allows for in-situ plasma cleaning and is well suited for producing highly ordered self-assembled monolayers (SAMs).
  • Hydrophilic and Hydrophobic Surfaces
  • The surface energy, or hydrophobicity of a surface, can be evaluated or measured by measuring a water contact angle. Water contact angle is the angle between the drop surface and a solid surface where a water droplet meets the solid surface. A surface with a water contact angle of smaller than 90°, the solid surface can be considered hydrophilic or polar. A surface with a water contact angle of greater than 90°, the solid surface can be considered hydrophobic or apolar.
  • Surface characteristics of coated surfaces can be adjusted in various ways suitable for oligonucleotide synthesis. In some cases, the surface is selected to be inert to the conditions of ordinary oligonucleotide synthesis; e.g. the solid surface may be devoid of free hydroxyl, amino, or carboxyl groups to the bulk solvent interface during monomer addition, depending on the selected chemistry. In some cases, the surface may comprise reactive moieties prior to the start of a first cycle, or first few cycles of an oligonucleotide synthesis process, wherein the reactive moieties can be quickly depleted to unmeasurable densities after one, two, three, four, five, or more cycles of the oligonucleotide synthesis reaction. The surface can further be optimized for well or pore wetting, e.g., by common organic solvents such as acetonitrile and the glycol ethers or aqueous solvents, relative to surrounding surfaces.
  • Without being bound by theory, the wetting phenomenon is understood to be a measure of the surface tension or attractive forces between molecules at a solid-liquid interface, and is expressed in dynes/cm2. For example, fluorocarbons have very low surface tension, which is typically attributed to the unique polarity (electronegativity) of the carbon-fluorine bond. In tightly structured Langmuir-Blodgett type films, surface tension of a layer can be primarily determined by the percent of fluorine in the terminus of the alkyl chains. For tightly ordered films, a single terminal trifluoromethyl group can render a surface nearly as lipophobic as a perfluoroalkyl layer. When fluorocarbons are covalently attached to an underlying derivatized solid (e.g. a highly crosslinked polymeric) support, the density of reactive sites can be lower than Langmuir-Blodgett and group density. For example, surface tension of a methyltrimethoxysilane surface can be about 22.5 mN/m and aminopropyltriethoxysilane surface can be about 35 mN/m. Briefly, hydrophilic behavior of surfaces is generally considered to occur when critical surface tensions are greater than 45 mN/m. As the critical surface tension increases, the expected decrease in contact angle is accompanied with stronger adsorptive behavior. Hydrophobic behavior of surfaces is generally considered to occur when critical surface tensions are less than 35 mN/m. At first, the decrease in critical surface tension is associated with oleophilic behavior, i.e. the wetting of the surfaces by hydrocarbon oils. As the critical surface tensions decrease below 20 mN/m, the surfaces resist wetting by hydrocarbon oils and are considered both oleophobic as well as hydrophobic. For example, silane surface modification can be used to generate a broad range of critical surface tensions. Accordingly, the methods and compositions of the invention may use surface coatings, e.g. those involving silanes, to achieve surface tensions of less than 5, 6, 7, 8, 9, 10, 12, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 110, 115, 120 mN/m, or higher. Further, the methods and compositions of the invention may use surface coatings, e.g. those involving silanes, to achieve surface tensions of more than 115, 110, 100, 90, 80, 70, 60, 50, 45, 40, 35, 30, 25, 20, 15, 12, 10, 9, 8, 7, 6 mN/m or less. The water contact angle and the surface tension of non-limiting examples of surface coatings, e.g., those involving silanes, are described in Table 1 and Table 2 of Arkles et al. (Silanes and Other Coupling Agents, Vol. 5v: The Role of Polarity in the Structure of Silanes Employed in Surface Modification. 2009), which is incorporated herein by reference in its entirety. The tables are replicated below.
  • TABLE 1
    Contact angles of water (degrees) on smooth surfaces
    Heptadecafluorodecyltrimethoxysilane 113-115
    Poly(tetrafluoroethylene) 108-112
    Polypropylene 108
    Octadecyldimethylchlorosilane 110
    Octadecyltrichlorosilane 102-109
    Tris(trimethylsiloxy)silylethyldimethylchlorosilane 103-104
    Octyldimethylchlorosilane 104
    Butyldimethylchlorosilane 100
    Trimethylchlorosilane 90-100
    Polyethylene 88-103
    Polystyrene 94
    Poly(chlorotrifluoroethylene) 90
    Human skin 75-90
    Diamond 87
    Graphite 86
    Silicon (etched) 86-88
    Talc 82-90
    Chitosan 80-81
    Steel 70-75
    Methoxyethoxyundecyltrichlorosilane 73-74
    Methacryloxypropyltrimethoxysilane 70
    Gold, typical (see gold, clean) 66
    Intestinal mucosa 50-60
    Kaolin 42-46
    Platinum 40
    Silicon nitride 28-30
    Silver iodide 17
    [Methoxy (polyethyleneoxy)propyl]trimethoxysilane 15-16
    Sodalime glass <15
    Gold, clean <10
    Trimethoxysilylpropyl substituted <10
    poly(ethyleneimine), hydrochloride
  • TABLE 2
    Critical surface tensions (mN/m)
    Heptadecafluorodecyltrichlorosilane 12
    Poly (tetrafluoroethylene) 18.5
    Octadecyltrichlorosilane 20-24
    Methyltrimethoxysilane 22.5
    Nonafluorohexyltrimethoxysilane 23
    Vinyltriethoxysilane 25
    Paraffin wax 25.5
    Ethyltrimethoxysilane 27.0
    Propyltrimethoxysilane 28.5
    Glass, sodalime (wet) 30.0
    Poly (chlorotrifluoroethylene) 31.0
    Polypropylene 31.0
    Poly (propylene oxide) 32
    Polyethylene 33.0
    Trifluoropropyltrimethoxysilane 33.5
    3-(2-Aminoethyl)aminopropyltrimethoxysilane 33.5
    Polystyrene 34
    p-Tolyltrimethoxysilane 34
    Cyanoethyltrimethoxysilane 34
    Aminopropyltriethoxysilane 35
    Acetoxy propyltrimethoxysilane 37.5
    Poly(methyl methacrylate) 39
    Poly(vinyl chloride) 39
    Phenyltrimethoxysilane 40.0
    Critical surface tensions (mN/m)
    Chloropropyltrimethoxysilane 40.5
    Mercaptopropyltrimethoxysilane 41
    Glycidoxypropyltrimethoxysilane 42.5
    Poly(ethylene terephthalate) 43
    Copper (dry) 44
    Poly(ethylene oxide) 43-45
    Aluminum (dry) 45
    Nylon 6/6 45-46
    Iron (dry) 46
    Glass, sodalime (dry) 47
    Titanium oxide (anatase) 91
    Ferric oxide 107
    Tin oxide 111
  • The surface of the substrate or a portion of the surface of the substrate can be functionalized or modified to be more hydrophilic or hydrophobic as compared to the surface or the portion of the surface prior to the functionalization or modification. In some cases, one or more surfaces can be modified to have a difference in water contact angle of greater than 90°, 85°, 80°, 75°, 70°, 65°, 60°, 55°, 50°, 45°, 40°, 35°, 30°, 25°, 20°, 15° or 10° as measured on one or more uncurved, smooth or planar equivalent surfaces. In some cases, the surface of the microstructures, channels, resolved loci, resolved reactor caps or other parts of the substrate may be modified to have a differential hydrophobicity corresponding to a difference in water contact angle that is greater than 90°, 85°, 80°, 75°, 70°, 65°, 60°, 55°, 50°, 45°, 40°, 35°, 30°, 25°, 20°, 15° or 10° as measured on uncurved, smooth or planar equivalent surfaces of such structures. Unless otherwise stated, water contact angles mentioned herein correspond to measurements that would be taken on uncurved, smooth or planar equivalents of the surfaces in question.
  • In some cases, hydrophilic resolved loci can be generated by first applying a protectant, or resist, over each loci within the substrate. The unprotected area can be then coated with a hydrophobic agent to yield an unreactive surface. For example, a hydrophobic coating can be created by chemical vapor deposition of (tridecafluorotetrahydrooctyl)-triethoxysilane onto the exposed oxide surrounding the protected channels or wells. Finally, the protectant, or resist, can be removed exposing the loci regions of the substrate for further modification and oligonucleotide synthesis. In some instances, the initial modification of such unprotected regions may resist further modification and retain their surface functionalization, while newly unprotected areas can be subjected to subsequent modification steps.
  • Substrate Etching
  • A process for carving features out of a substrate (e.g., a silicon plate) may include providing a substrate having a device layer and a handle layer, wherein the device layer is optionally separated from the handle layer by an electrical insulator layer, e.g., a layer of silicon dioxide—an exemplary substrate is a SOI wafer. In some instances, the provided substrate is oxidized on both surfaces. Photolithography may be applied to the device side of the substrate to create a mask of photoresist. In a subsequent step, a deep reactive-ion etching (DRIE) step may be used to etch vertical side-walls (e.g., until an insulator layer in a substrate comprising an insulator layer) at locations devoid of the photoresist. In a following step, the photoresist may be stripped. In some instances, photolithography, DRIE and photoresist strip steps are repeated on the substrate handle side. In cases wherein the substrate comprises an insulator layer such as silicon dioxide, buried oxide (BOX) may be removed using an etching process. Thermal oxidation can then be applied to remove contaminating polymers that may have been deposited on the side walls during the method. In a subsequent step, the thermal oxidation may be stripped using a wet etching process. In some instances, this method is used to generate a substrate having the features of a substrate exemplified in FIGS. 4A-4B. As another example, the process for manufacturing a substrate comprises a front-end process method comprising providing a starting material substrate, oxidizing both the device and handle sides; performing photolithography, DRIE and stripping of photoresist on the handle side; performing photolithography, DRIE and stripping of photoresist on the device side; removal of the oxide layer (e.g., BOX); and oxide growth (e.g., oxide is coated on one or more surfaces of the substrate to create a silicon substrate having a plurality of features).
  • In some instances, the substrate starting material comprises silicon. In some instances, the substrate is oxidized on one or more surfaces. In some instances, photolithography is applied to the front-side, back-side or both the front and back sides of the substrate, for example, to the back-side, to create a mask of photoresist. As a next step, the substrate is etched at locations devoid of photoresist, in many cases, beyond the oxidized layer, to create wells. As an example, the back-side is etched. In a subsequent step, the photoresist is stripped. In some instances, wherein photolithography was first applied to one side of the substrate, following photoresist stripping, a second side of the substrate is subjected to photolithography. For example, the back-side was first subjected to photolithography followed by photolithography of the front-side of the substrate. In some examples, a deep reactive-ion etching (DRIE) is used to etch vertical side walls to a prescribed depth, for example, about 450 um. In some cases, DRIE is used on the front-side, back-side or both the front and back sides during photolithography. In some instances, only one side of a substrate is etched to create three-dimensional features. In some instances, two sides, e.g., device and handle sides, of a substrate is etched to create three-dimensional features. In some processes, as an alternative or supplement to etching by DRIE, a SOI substrate (silicon on insulator silicon wafer) is used and the handle layer is etched down to the buried oxide, wherein the buried oxide can serve as an etch stop. Following photolithography on a second side of a substrate, the photoresist is stripped to generate a desired pattern. For example, the front-end is resist stripped to generate three dimensional features. In some cases, contaminates of the process (e.g., fluoropolymers) are removed by thermal oxidation followed by stripping of the thermal oxidation by a wet etching process. The substrate processed may comprise a plurality of wells, where the distance from the center of one main channel to the center of another main channel is about 1.69 mm and the total height of the main channel/microchannel is about 450 um.
  • Fiducial Marks
  • A substrate described herein may comprise fiducial marks to facilitate alignment with other components of a system. In some cases, substrates have one or more fiducial marks, e.g. 2, 3, 4, 5, 6, 7, 8, 9, 10, or more fiducial marks. A fiducial mark may be located near the origin, where the fiducial mark is closer to the origin than any one cluster. In some instances, a fiducial mark is located near an edge of the substrate portion. The fiducial mark may be located from about 0.1 mm to about 10 mm from the edge of the substrate portion, e.g., about 0.5 mm from the edge. The fiducial mark may be located close to or distant to a cluster. For example, a fiducial is located from about 1 mm to about 10 mm form a cluster, e.g., 1.69 mm. In some instances, a distance from the center of a fiducial mark and a nearest corner of a substrate in one dimension is from about 0.5 mm to about 10 mm, e.g., about 1 mm. In some instances, a length of a fiducial mark in one dimension is from about 0.5 mm to about 5 mm, e.g., about 1 mm. In some instances, the width of a fiducial mark is from about 0.01 mm to about 2 mm, e.g., 0.05 mm. The substrates described herein, in some instances, comprise one or more regions for annotation. In some instances, a substrate may have a label or serial number which is located a distance (e.g., 4 mm) from the edge of the substrate with a length (e.g., 9 mm) and width (e.g., 1.5 mm).
  • Oligonucleic Acid Synthesis
  • Structures having modified surfaces described herein may be used for de novo synthesis processes. An exemplary workflow for one such process is divided generally into phases: (1) de novo synthesis of a single stranded oligonucleic acid library, (2) joining oligonucleic acids to form larger fragments, (3) error correction, (4) quality control, and (5) shipment, FIG. 12 . Prior to de novo synthesis, an intended nucleic acid sequence or group of nucleic acid sequences is preselected. For example, a group of genes is preselected for generation.
  • Once preselected nucleic acids for generation are selected, a predetermined library of oligonucleic acids is designed for de novo synthesis. Various suitable methods are known for generating high density oligonucleic acid arrays. In the workflow example, a surface layer 1201 is provided. In the example, chemistry of the surface is altered in order to improve the oligonucleic acid synthesis process. Areas of low surface energy are generated to repel liquid while areas of high surface energy are generated to attract liquids. The surface itself may be in the form of a planar surface or contain variations in shape, such as protrusions, microwells, or microchannels which increase surface area. In the workflow example, high surface energy molecules selected serve a dual function of supporting DNA chemistry, as described in International Patent Application Publication WO/2015/021280, which is herein incorporated by reference in its entirety.
  • In situ preparation of oligonucleic acid arrays is generated on a solid support and utilizes single nucleotide extension process to extend multiple oligomers in parallel. A device, such as an oligonucleic acid synthesizer, is designed to release reagents in a step wise fashion such that multiple oligonucleic acids extend, in parallel, one residue at a time to generate oligomers with a predetermined nucleic acid sequence 1202. In some cases, oligonucleic acids are cleaved from the surface at this stage. Cleavage may include gas cleavage, e.g., with ammonia or methylamine.
  • The generated oligonucleic acid libraries are placed in a reaction chamber. In this exemplary workflow, the reaction chamber (also referred to as “nanoreactor”) is a silicon coated main channel, containing PCR reagents and lowered onto the oligonucleic acid library 1203. Prior to or after the sealing 1204 of the oligonucleic acids, a reagent is added to release the oligonucleic acids from the surface. In the exemplary workflow, the oligonucleic acids are released subsequent to sealing of the nanoreactor 1205. Once released, fragments of single stranded oligonucleic acids hybridize in order to span an entire long range sequence of DNA. Partial hybridization 1205 is possible because each synthesized oligonucleic acid is designed to have a small portion overlapping with at least one other oligonucleic acid in the pool.
  • After hybridization, a PCA reaction is commenced. During the polymerase cycles, the oligonucleic acids anneal to complementary fragments and gaps are filled in by a polymerase. Each cycle increases the length of various fragments randomly depending on which oligonucleic acids find each other. Complementarity amongst the fragments allows for forming a complete large span of double stranded DNA 1206.
  • After PCA is complete, the nanoreactor is separated from the surface 1207 and positioned for interaction with a polymerase 1208. After sealing, the nanoreactor is subject to PCR 1209 and the larger nucleic acids are formed. After PCR 1210, the nanochamber is opened 1211, error correction reagents are added 1212, the chamber is sealed 1213 and an error correction reaction occurs to remove mismatched base pairs and/or strands with poor complementarity from the double stranded PCR amplification products 1214. The nanoreactor is opened and separated 1215. Error corrected product is next subject to additional processing steps, such as PCR and molecular bar coding, and then packaged 1222 for shipment 1223.
  • In some cases, quality control measures are taken. After error correction, quality control steps include for example interaction with a wafer having sequencing primers for amplification of the error corrected product 1216, sealing the wafer to a chamber containing error corrected amplification product 1217, and performing an additional round of amplification 1218. The nanoreactor is opened 1219 and the products are pooled 1220 and sequenced 1221. After an acceptable quality control determination is made, the packaged product 1222 is approved for shipment 1223.
  • Oligonucleic acids may be synthesized on a substrate described herein using a system comprising an oligonucleic acid synthesizer that deposits reagents necessary for synthesis. Reagents for oligonucleic acid synthesis include, for example, reagents for oligonucleic acid extension and wash buffers. As non-limiting examples, the oligonucleic acid synthesizer deposits coupling reagents, capping reagents, oxidizers, de-blocking agents, acetonitrile and gases such as nitrogen gas. In addition, the oligonucleic acid synthesizer optionally deposits reagents for the preparation and/or maintenance of substrate integrity. An oligonucleic acid synthesizer may comprise material deposition devices that can move in the X-Y direction to align with the location of the substrate. The oligonucleic acid synthesizer can also move in the Z direction to seal with the substrate, forming a resolved reactor. In some instances, a substrate having a plurality of clusters is configured to seal with a capping element having a plurality of caps, wherein when the substrate and capping element are sealed, each cluster is separate from another cluster to form separate resolved reactors for each cluster. In some instances, the capping element is not present in the system or is present and stationary. A resolved reactor is configured to allow for the transfer of fluid, including oligonucleic acids and/or reagents, from the substrate to the capping element and/or vice versa. Fluid may pass through either or both the substrate and the capping element and includes, without limitation, coupling reagents, capping reagents, oxidizers, de-blocking agents, acetonitrile and nitrogen gas. The oligonucleic acid synthesizer of an oligonucleic acid synthesis system may comprise a plurality of material deposition devices, for example from about 1 to about 50 material deposition devices. Each material deposition device, in various instances, deposits a reagent component that is different from another material deposition device. In some cases, each material deposition device has a plurality of nozzles, where each nozzle is optionally configured to correspond to a cluster on a substrate. For example, for a substrate having 256 clusters, a material deposition device has 256 nozzles and 100 μm fly height. In some cases, each nozzle deposits a reagent component that is different from another nozzle.
  • The substrates described herein comprise actively functionalized surfaces configured to support the attachment and synthesis of oligonucleic acids. Synthesized oligonucleic acids include oligonucleic acids comprising modified and/or non-canonical bases and/or modified backbones. In various methods, a library of oligonucleic acids having pre-selected sequences is synthesized on a substrate. In some cases, one or more of the oligonucleic acids has a different sequence and/or length than another oligonucleic acid in the library. In some instances, the stoichiometry of each oligonucleic acid synthesized on a substrate is controlled and tunable by varying one or more features of the substrate (e.g., functionalized surface) and/or oligonucleic acid sequence to be synthesized; one or more methods for substrate functionalization and/or oligonucleic acid synthesis; or a combination thereof. In many instances, controlling the density of a growing oligonucleic acid on a resolved locus of a substrate allows for oligonucleic acids to be synthesized with a low error rate.
  • An example of a synthesis method that is useful with the substrates provided herein is one based on phosphoramidite chemistry. In some instances, oligonucleic acid synthesis methods comprise coupling a linker to a surface of a substrate, for example, to an actively functionalized surface of a substrate. In some instances, a linker separates a synthesized oligonucleic acid from a surface of the substrate. A linker includes a cleavable linker, such as a photocleavable linker. In some instances, a synthesized oligonucleic acid comprises a cleavable moiety that is introduced during synthesis. In some cases, a synthesized oligonucleic acid does not comprise a linker. For example, the synthesized oligonucleic acid is separated from the linker by one or more cleavable moieties. In some instances, the synthesized oligonucleic acid comprises a primer and/or adapter sequence that connects to a linker.
  • Without wishing to be bound by theory, the distance between extending oligonucleic acids is a factor correlating to error rate occurrence in synthesis of an oligonucleic acid library. One way to reduce the frequency of error is to minimize chain interaction during extension. Polymer “wobble” is controlled by altering the length of the tethering group at the base of the extending polymeric structure. In some instances, regulating “wobble” reduces error rate of polymer over the course of the synthesis process. In some instances, a linker comprises one or more bases coupled to the surface of a substrate and a cleavable moiety, wherein the cleavable moiety is configured to connect to the synthesized oligonucleic acid. In some cases, a linker is referred to as a tether or a tether region. In some instances, a linker comprises about or at least about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50 or more bases located between a surface of a substrate and a synthesized oligonucleic acid. In some instances, a linker is synthesized and extends 12 to 25 bases from a device surface.
  • In some instances, a linker comprises a cleavable moiety, wherein the cleavable moiety is a modified or non-canonical base. When a plurality of synthesized oligonucleic acids of a library are connected to a substrate surface by a plurality of linkers having the same cleavable moiety, the cleavable moiety is referred to as a universal moiety. Examples of cleavable moieties include, without limitation, thymidine-succinyl hexamide CED phosphoramidite and DMMA. In some instances, a cleavable moiety is gas cleavable. In some instances, the linker comprises thymidine-succinyl hexamide CED phosphoramidite or DMMA. In some instances, the linker comprises a photocleavable primer. In an example, a photocleavable linker allows for the synthesized oligonucleic acid to be removed from the substrate without cleaving the protecting groups on the nitrogenous functionalities of each base, for example, by irradiation with light at about 350 nm.
  • Oligonucleic acids synthesized using the methods and/or substrates described herein comprise, in various instances, at least about 50, 60, 70, 75, 80, 90, 100, 120, 150, 200, 300, 400, 500, 600, 700, 800 or more bases. In some instances, a library of oligonucleic acids is synthesized, wherein a population of distinct oligonucleic acids are assembled to generate a larger nucleic acid comprising at least about 500; 1,000; 2,000; 3,000; 4,000; 5,000; 6,000; 7,000; 8,000; 9,000; 10,000; 11,000; 12,000; 13,000; 14,000; 15,000; 16,000; 17,000; 18,000; 19,000; 20,000; 25,000; 30,000; 40,000; or 50,000 bases. In some instances, oligonucleic acid synthesis methods described herein are useful for the generation of an oligonucleic acid library comprising at least 500; 1,000; 5,000; 10,000; 20,000; 50,000; 100,000; 200,000; 300,000; 400,000; 500,000; 600,000; 700,000; 800,000; 900,000; 1,000,000; 1,100,000; 1,200,000; 1,300,000; 1,400,000; 1,500,000; 1,600,000; 1,700,000; 1,800,000; 1,900,000; 2,000,000; 2,200,000; 2,400,000; 2,600,000; 2,800,000; 3,000,000; 3,500,000; 4,00,000; or 5,000,000 distinct oligonucleic acids. In some instances, at least about 1 μmol, 10 pmol, 20 pmol, 30 pmol, 40 pmol, 50 pmol, 60 pmol, 70 pmol, 80 pmol, 90 pmol, 100 pmol, 150 pmol, 200 pmol, 300 pmol, 400 pmol, 500 pmol, 600 pmol, 700 pmol, 800 pmol, 900 pmol, 1 nmol, 5 nmol, 10 nmol, 100 nmol or more of an oligonucleic acid is synthesized within a locus.
  • Methods for oligonucleic acid synthesis on a surface provided herein allow for synthesis at a fast rate. As an example, at least 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 35, 40, 45, 50, 55, 60, 70, 80, 90, 100, 125, 150, 175, 200 nucleotides per hour, or more are synthesized. Nucleotides include adenine, guanine, thymine, cytosine, uridine building blocks, or analogs/modified versions thereof. In some instances, libraries of oligonucleic acids are synthesized in parallel on substrate. For example, a substrate comprising about or at least about 100; 1,000; 10,000; 100,000; 1,000,000; 2,000,000; 3,000,000; 4,000,000; or 5,000,000 resolved loci is able to support the synthesis of at least the same number of distinct oligonucleic acids, wherein oligonucleic acid encoding a distinct sequence is synthesized on a resolved locus. In some instances, a library of oligonucleic acids are synthesized on a substrate with low error rates described herein in less than about three months, two months, one month, three weeks, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2 days, 24 hours, 18 hours, 12 hours or less. In some instances, larger nucleic acids assembled from an oligonucleic acid library synthesized with low error rate using the substrates and methods described herein are prepared in less than about three months, two months, one month, three weeks, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2 days, 24 hours, 18 hours, 12 hours or less. In some instances, up to about 800,000 distinct oligonucleic acids having sizes up to about 130 base pairs in length each are synthesized with an error rate below 1:1000, 1:2000, 1:3000 or less on a substrate described herein and using a method described herein in a span of less than about 24 hours.
  • In some instances, oligonucleic acid error rate is dependent on the efficiency of one or more chemical steps of oligonucleic acid synthesis. In some cases, oligonucleic acid synthesis comprises a phosphoramidite method, wherein a base of a growing oligonucleic acid chain is coupled to phosphoramidite. In some instances, coupling efficiency of the base is related to error rate. For example, higher coupling efficiency correlates to lower error rates. In some cases, the substrates and/or synthesis methods described herein allow for a coupling efficiency greater than 98%, 98.5%, 99%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9%, 99.95%, 99.96%, 99.97%, 99.98%, or 99.99%. In some cases, an oligonucleic acid synthesis method comprises a double coupling process, wherein a base of a growing oligonucleic acid chain is coupled with a phosphoramidite, the oligonucleic acid is washed and dried, and then treated a second time with a phosphoramidite. In some instances, efficiency of deblocking in a phosphoramidite oligonucleic acid synthesis method contributes to error rate. In some cases, the substrates and/or synthesis methods described herein allow for removal of 5′-hydroxyl protecting groups at efficiencies greater than 98%, 98.5%, 99%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9%, 99.95%, 99.96%, 99.97%, 99.98%, or 99.99%. In some instances, error rate is reduced by minimization of depurination side reactions.
  • Oligonucleic acids synthesized using the methods and/or substrates described herein encode for, in various instances, at least about 50, 60, 70, 75, 80, 90, 100, 120, 150, 200, 240, 300, 400, 500, 600, 700, 800, 900, 1,000, 6000, 6144, 10,000, or more genes. In some instances, a library of oligonucleic acids encode for at least 200 genes. In some instances, a library of oligonucleic acids encode for genes at least 500 bases, 1 kb, 2 kb, 3 kb, 4 kb, 5 kb or more in length.
  • Methods for oligonucleic acid synthesis, in various instances, include processes involving phosphoramidite chemistry. In some instances, oligonucleic acid synthesis comprises coupling a base with phosphoramidite. In some instances, oligonucleic acid synthesis comprises coupling a base by deposition of phosphoramidite under coupling conditions, wherein the same base is optionally deposited with phosphoramidite more than once, i.e., double coupling. In some instances, oligonucleic acid synthesis comprises capping of unreacted sites. In some cases, capping is optional. In some instances, oligonucleic acid synthesis comprises oxidation. In some instances, oligonucleic acid synthesis comprises deblocking or detritylation. In some instances, oligonucleic acid synthesis comprises sulfurization. In some cases, oligonucleic acid synthesis comprises either oxidation or sulfurization. In some instances, between one or each step during an oligonucleic acid synthesis reaction, the substrate is washed, for example, using tetrazole or acetonitrile. Time frames for any one step in a phosphoramidite synthesis method include less than about 2 min, 1 min, 50 sec, 40 sec, 30 sec, 20 sec and 10 sec.
  • Oligonucleic acid synthesis using a phosphoramidite method comprises the subsequent addition of a phosphoramidite building block (e.g., nucleoside phosphoramidite) to a growing oligonucleic acid chain for the formation of a phosphite triester linkage. Phosphoramidite oligonucleic acid synthesis proceeds in the 3′ to 5′ direction. Phosphoramidite oligonucleic acid synthesis allows for the controlled addition of one nucleotide to a growing nucleic acid chain per synthesis cycle. In some instances, each synthesis cycle comprises a coupling step. Phosphoramidite coupling involves the formation of a phosphite triester linkage between an activated nucleoside phosphoramidite and a nucleoside bound to the substrate, for example, via a linker. In some instances, the nucleoside phosphoramidite is provided to the substrate activated. In some instances, the nucleoside phosphoramidite is provided to the substrate with an activator. In some instances, nucleoside phosphoramidites are provided to the substrate in a 1.5, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100-fold excess or more over the substrate-bound nucleosides. In some instances, the addition of nucleoside phosphoramidite is performed in an anhydrous environment, for example, in anhydrous acetonitrile. Following addition of a nucleoside phosphoramidite, the substrate is optionally washed. In some instances, the coupling step is repeated one or more additional times, optionally with a wash step between nucleoside phosphoramidite additions to the substrate. In some instances, an oligonucleic acid synthesis method used herein comprises 1, 2, 3 or more sequential coupling steps. Prior to coupling, in many cases, the nucleoside bound to the substrate is de-protected by removal of a protecting group, where the protecting group functions to prevent polymerization. A common protecting group is 4,4′-dimethoxytrityl (DMT).
  • Following coupling, phosphoramidite oligonucleic acid synthesis methods optionally comprise a capping step. In a capping step, the growing oligonucleic acid is treated with a capping agent. A capping step is useful to block unreacted substrate-bound 5′—OH groups after coupling from further chain elongation, preventing the formation of oligonucleic acids with internal base deletions. Further, phosphoramidites activated with 1H-tetrazole may react, to a small extent, with the O6 position of guanosine. Without being bound by theory, upon oxidation with I2/water, this side product, possibly via O6-N7 migration, may undergo depurination. The apurinic sites may end up being cleaved in the course of the final deprotection of the oligonucleotide thus reducing the yield of the full-length product. The O6 modifications may be removed by treatment with the capping reagent prior to oxidation with I2/water. In some instances, inclusion of a capping step during oligonucleic acid synthesis decreases the error rate as compared to synthesis without capping. As an example, the capping step comprises treating the substrate-bound oligonucleic acid with a mixture of acetic anhydride and 1-methylimidazole. Following a capping step, the substrate is optionally washed.
  • In some instances, following addition of a nucleoside phosphoramidite, and optionally after capping and one or more wash steps, the substrate bound growing nucleic acid is oxidized. The oxidation step comprises the phosphite triester is oxidized into a tetracoordinated phosphate triester, a protected precursor of the naturally occurring phosphate diester internucleoside linkage. In some cases, oxidation of the growing oligonucleic acid is achieved by treatment with iodine and water, optionally in the presence of a weak base (e.g., pyridine, lutidine, collidine). Oxidation may be carried out under anhydrous conditions. Oxidation may be carried out using, for example, tert-Butyl hydroperoxide or (1S)-(+)-(10-camphorsulfonyl)-oxaziridine (CSO). In some methods, a capping step is performed following oxidation. A second capping step allows for substrate drying, as residual water from oxidation that may persist can inhibit subsequent coupling. Following oxidation, the substrate and growing oligonucleic acid is optionally washed. In some instances, the step of oxidation is substituted with a sulfurization step to obtain oligonucleotide phosphorothioates, wherein any capping steps can be performed after the sulfurization. Many reagents are capable of the efficient sulfur transfer, including but not limited to, 3-(dimethylaminomethylidene)amino)-3H-1,2,4-dithiazole-3-thione, DDTT, 3H-1,2-benzodithiol-3-one 1,1-dioxide, also known as Beaucage reagent, and N,N,N′N′-tetraethylthiuram disulfide (TETD).
  • In order for a subsequent cycle of nucleoside incorporation to occur through coupling, the protected 5′ end of the substrate bound growing oligonucleic acid must be removed so that the primary hydroxyl group can react with a next nucleoside phosphoramidite. In some instances, the protecting group is DMT and deblocking occurs with trichloroacetic acid in dichloromethane. Conducting detritylation for an extended time or with stronger than recommended solutions of acids may lead to increased depurination of solid support-bound oligonucleotide and thus reduces the yield of the desired full-length product. Methods and compositions of the invention described herein provide for controlled deblocking conditions limiting undesired depurination reactions. In some cases, the substrate bound oligonucleic acid is washed after deblocking. In some cases, efficient washing after deblocking contributes to synthesized oligonucleic acids having a low error rate.
  • Methods for the synthesis of oligonucleic acids typically involve an iterating sequence of the following steps: application of a protected monomer to an actively functionalized surface (e.g., locus) to link with either the activated surface, a linker or with a previously deprotected monomer; deprotection of the applied monomer so that it can react with a subsequently applied protected monomer; and application of another protected monomer for linking. One or more intermediate steps include oxidation or sulfurization. In some cases, one or more wash steps precede or follow one or all of the steps.
  • Methods for phosphoramidite based oligonucleic acid synthesis comprise a series of chemical steps. In some instances, one or more steps of a synthesis method involve reagent cycling, where one or more steps of the method comprise application to the substrate of a reagent useful for the step. For example, reagents are cycled by a series of liquid deposition and vacuum drying steps. For substrates comprising three-dimensional features such as wells, microwells, channels, microchannels and the like, reagents are optionally passed through one or more regions of the substrate via the wells and/or channels. In some instances, reagents are passed through the substrate during synthesis. In some cases, reagents are passed horizontally through the substrate. In some cases, reagents are passed vertically through the substrate. In some instances, reagents are passed over a substrate having curved features to enhance flow. In some cases, reagents are delivered to the substrate through the use of photoresist. In some instances, reagents are delivered to the substrate without moving the substrate. For example, reagents are passed over resolved loci within the substrate by flowing them through the substrate from one surface to an opposite surface of the substrate. In some instances, the substrate is moved, for example, to a flow cell, for reagent application, where it is then optionally repositioned. In an example, the substrate is deposited with a nucleoside using an oligonucleic acid synthesizer, moved to a flow cell for treating the substrate to one or more select reagents, and then repositioned back to the oligonucleic acid synthesizer for deposition of a subsequent monomer. Reagent delivery approaches suitable for the synthesis methods of the disclosure include manual and automatic, including use of robotic devices and pulse jets. Reagents include any component of an oligonucleic acid synthesis method, including chemical moieties such as nucleosides, washing solutions, and gases such as nitrogen.
  • In some instances, one or more reagents applied to the surface of a substrate during oligonucleic acid synthesis comprise a solvent. In some cases, a solvent comprises propylene carbonate. In some cases, a solvent comprises 2-methylglutaronitrile and/or 3-methoxypropionitrile. In some cases, a solvent comprises glutaronitrile. In some cases, a solvent comprises adiponitrile. In some instances, the solvent allows for high surface tension for reagent deposition. In some instances, the solvent allows for low surface tension for reagent deposition.
  • In some instances, the volume of reagents applied to a surface of substrate during oligonucleic acid synthesis is selected on the size, location and/or density of the surface to which the reagent is applied (e.g., an actively functionalized locus). In some instances, the volume of a drop of reagent applied to a surface during oligonucleic acid synthesis (e.g., deposition of a nucleoside) is less than about 0.5 picoliters (pL), 1 pL, 5 pL, 10 pL, 50 pL, 100 pL, 500 pL, 1000 pL, 5000 pL, 10000 pL, 100000 pL, 1000000 pL or 10000000 pL. In some instances, the reagents are delivered in droplets that have a total volume of about 47 pL or less. In some instances, the reagents are delivered in droplets that have a total volume of about 30 to 50 pL. In some instances, the reagents are delivered in droplets that have a total volume of about 50, 49, 48, 47, 46, 44, 45, 43, 42, 41, 40, 39, 38, 37, 36, 35, 34, 33, 32, 31, 30, 29, 28, 27, 26, or 25 pL. In some instances, the rate at which a drop of reagent is applied is at least about 0.5, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 30, 50 or 100 m/sec.
  • Oligonucleic acid synthesis methods include methods for the application of reagents during one or more steps during synthesis. Controlled application of reagents, such as nucleoside monomers to distinct regions of a substrate is important to achieve low error rates. In some instances, a reagent is deposited directly into a microchannel, with little or no contamination to an adjacent microchannel. In some cases, the volume of a reagent to be deposited within a three-dimensional feature such as a well or channel is adjusted to a small enough size to minimize cross-contamination. In some instances, the reagents are delivered in droplets that have a diameter of less than about 5, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190 or 200 um. A non-limiting method to reduce cross-contamination includes bringing the device that deposits the reagent sufficiently close to the surface such the deposited droplet falls substantially within the selected feature.
  • In some instances, efficient washing to remove unincorporated nucleosides contributes to low error rate. In some instances, the composition of the wash contributes to low error rate. As described herein, washing during oligonucleic acid synthesis includes one or all wash steps performed during oligonucleic acid synthesis. In some cases, a wash step is performed wherein at least or about 60%, 70, 80, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99 or 100% of unincorporated nucleosides or extension reaction reagent are removed from the surface of the substrate. In some cases, a wash step is performed wherein at least or about 95.0, 95.1, 95.2, 95.3, 95.4, 95.5, 95.6, 95.7, 95.8, 95.9, 96.0, 96.1, 96.2, 96.3, 96.4, 96.5, 96.6, 96.7, 96.8, 96.9, 97.0, 97.1, 97.2, 97.3, 97.4, 97.5, 97.6, 97.7, 97.8, 97.9, 98.0, 98.1, 98.2, 98.3, 98.4, 98.5, 98.6, 98.7, 98.8, 98.9, 99.0, 99.1, 99.2, 99.3, 99.4, 99.5, 99.6, 99.7, 99.8, 99.9, or 100% of unincorporated nucleosides or extension reaction reagent are removed from the surface of the substrate. In some cases, the configuration of a substrate such as one with narrow microchannels, contributes to wash efficiency and error rate. In some cases, substrates having channels are washed by passage of a wash solution through the substrate, minimizing fluid contact of the growing oligonucleic acids. In some cases, the geometry of fluid flow during washing controls interfacial instability. For example, a substrate that is substantial planar, or two-dimensional, may have a curved surface to enhance wash efficiency and therefore error rate. In some instances, optimized wash conditions include those that minimize contact time between the wash reagent and the growing oligonucleic acid. For example, the passage of a wash solution through three-dimensional features allows for the effect washing of all surfaces in a short period of time. A water contact angle for the substrate, in particular, for regions of synthesis and/or surrounding areas, may be chosen in order to reduce depurination and/or speed of synthesis. In some instances, lower amount of depurination may be achieved on surfaces of higher surface energy, i.e. lower contact angle. For example, depurination occurs at a rate less than 0.1%, 0.05%, 0.01%, 0.005%, 0.001%, 0.0005%, or 0.0001%.
  • In some instances, the surface properties of the substrate change during oligonucleic acid synthesis. Typically, the substrate at the beginning of synthesis can be relatively hydrophobic and while synthesis proceeds, may become increasingly hydrophilic. Oligonucleic acid features can gain substantial surface energy with increasing oligonucleotide length. Generally, these sites or features consisting of protected oligonucleotide acquire enough surface energy to become spontaneously wet to high surface tension organic solvents commonly used in phosphoramidite synthesis, such as acetonitrile or propylene carbonate, after about 10-20 synthesis cycles. The methods and compositions described allow for varying parameters, such as time, flow rate, temperature, volume, viscosity, and/or reagent concentration, during the synthesis of a growing oligonucleic acid as a function of length to account for the changing surface properties on loci of the surface. Such a variation may be applied by changing parameters in constant or varying increments at repeating cycles of the synthesis. Alternatively, parameters may be changed at only selected cycles of the synthesis and can optionally follow a pattern, such as every other cycle, every third, fourth, fifth, sixth, seventh, eighth, ninth, tenth cycle etc.
  • Oligonucleic acid synthesis methods described herein are suitable for the spatial control of oligonucleic acid synthesis within a small area of a substrate, e.g., a locus. In some instances, oligonucleic acid methods comprise phosphoramidite chemistry. In some instances, spatial control of oligonucleic acid synthesis is achieved using an oligonucleic acid synthesizer. In some instances, spatial control of oligonucleic acid synthesis is achieved using physical masks. In some instances, spatial control of oligonucleic acid synthesis is achieved by modulation of a 5′ hydroxyl deblocking during phosphoramidite synthesis. In some instances, spatial control of oligonucleic acid synthesis is achieved by photolithographic deprotection of photolabile monomers. In some instances, spatial control of oligonucleic acid synthesis is achieved by digital activation of photogenerated acids to carry out standard detritylation.
  • In some instances, the surface of the substrate that provides support for oligonucleic acid synthesis is chemically modified to allow for the synthesized oligonucleic acid chain to be cleaved from the surface. In some cases, the oligonucleic acid chain is cleaved at the same time as the oligonucleic acid is deprotected. In some cases, the oligonucleic acid chain is cleaved after the oligonucleic acid is deprotected. In an exemplary scheme, a trialkoxysilyl amine (e.g., (CH3CH2O)3Si—(CH2)2—NH2) is reacted with surface SiOH groups of a substrate, followed by reaction with succinic anhydride with the amine to create and amide linkage and a free OH on which the nucleic acid chain growth is supported.
  • In some instances, oligonucleic acids are synthesized with photolabile protecting groups, where the hydroxyl groups generated on the surface are blocked by photolabile-protecting groups. When the surface is exposed to UV light, e.g., through a photolithographic mask, a pattern of free hydroxyl groups on the surface may be generated. These hydroxyl groups can react with photoprotected nucleoside phosphoramidites, according to phosphoramidite chemistry. A second photolithographic mask can be applied and the surface can be exposed to UV light to generate second pattern of hydroxyl groups, followed by coupling with 5′-photoprotected nucleoside phosphoramidite. Likewise, patterns can be generated and oligomer chains can be extended. Without being bound by theory, the lability of a photocleavable group depends on the wavelength and polarity of a solvent employed and the rate of photocleavage may be affected by the duration of exposure and the intensity of light. This method can leverage a number of factors, e.g., accuracy in alignment of the masks, efficiency of removal of photo-protecting groups, and the yields of the phosphoramidite coupling step. Further, unintended leakage of light into neighboring sites can be minimized. The density of synthesized oligomer per spot can be monitored by adjusting loading of the leader nucleoside on the surface of synthesis.
  • Oligonucleotide Libraries with Low Error Rates
  • The term “error rate” may also be referred to herein as a comparison of the collective sequence encoded by oligonucleic acids generated compared to the aggregate sequence of a predetermined longer nucleic acid, e.g., a gene. Oligonucleic acids are synthesized on a substrate described herein in a process that minimizes the error rate. For example, error rate is less than 1 in 500 bases, 1 in 1000 bases, 1 in 1500 bases, 1 in 2000 bases, 1 in 2500 bases, 1 in 3000, 1 in 5000 bases or less. In some instances, low error rates are achieved for synthesized oligonucleic acid libraries having at least 20,000; 40,000; 60,000; 80,000; 100,000; 200,000; 300,000; 400,000; 500,000; 600,000; 700,000; 800,000; 1,000,000; or 2,000,000 or more oligonucleic acids. In some cases, a subset of oligonucleic acids in the library has the same sequence. In some cases, one or more of the oligonucleic acids in the library comprises a different sequence. Error rates include mismatch error rate, deletion error rate, insertion error rate, indel error rate, and any combination thereof.
  • In some instances, low overall error rate or low error rates for individual types of errors are achieved. Individual types of error rates include deletions, insertions, or substitutions for an oligonucleic acid library synthesized on the substrate. In some instances, oligonucleic acids synthesized on the substrate have an average error rate of about 1:500, 1:1000, 1:2000, 1:3000, 1:4000, 1:5000, 1:6000, 1:7000, 1:8000, 1:9000, 1:10000, 1:20000, 1:30000, 1:40000, 1:50000 or less. In some instances, these error rates are for at least 50%, 60%, 70%, 80%, 90%, 95%, 98%, 99%, 99.5%, or more of the oligonucleic acids synthesized. In some instances, these error rates are for 100% of the oligonucleic acids synthesized.
  • In some instances, oligonucleic acids synthesized on the substrate have an average deletion error rate of about 1:500, 1:1000, 1:2000, 1:3000, 1:4000, 1:5000, 1:6000, 1:7000, 1:8000, 1:9000, 1:10000, 1:20000, 1:30000, 1:40000, 1:50000 or less. In some instances, oligonucleic acids synthesized on the substrate have an average insertion error rate of about 1:500, 1:1000, 1:2000, 1:3000, 1:4000, 1:5000, 1:6000, 1:7000, 1:8000, 1:9000, 1:10000, 1:20000, 1:30000, 1:40000, 1:50000 or less. In some instances, oligonucleic acids synthesized on the substrate have an average substitution error rate of about 1:500, 1:1000, 1:2000, 1:3000, 1:4000, 1:5000, 1:6000, 1:7000, 1:8000, 1:9000, 1:10000, 1:20000, 1:30000, 1:40000, 1:50000 or less. In some instances, overall error rate or error rates for individual types of errors such as deletions, insertions, or substitutions for each oligonucleotide synthesized on the substrate, for at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 98%, 99%, 99.5%, or more of the oligonucleotides synthesized on the substrate, or the substrate average may fall between about 1:500 and 1:50000, 1:500 and 1:40000; 1:500 and 1:30000; 1:500 and 1:20000; 1:500 and 1:10000; 1:500 and 1:9000; 1:500 and 1:8000; 1:500 and 1:7000; 1:500 and 1:6000; or 1:500 and 1:5000.
  • In some instances, the methods and systems described herein for oligonucleic acid synthesis results in minimal synthesis of truncation products that are less than the full length of the predetermined oligonucleic acid sequence. In some cases, a library of oligonucleic acids are synthesized with less than 1%, 0.5%, 0.1%, 0.05%, 0.01%, 0.005%, 0.001%, or 0.0001% comprising truncation products. In some instances, the methods and systems described herein for oligonucleic acid synthesis result in minimal synthesis of products that are greater than predetermined oligonucleic acid sequence length. In some cases, a library of oligonucleic acids are synthesized with less than 1%, 0.5%, 0.1%, 0.05%, 0.01%, 0.005%, 0.001%, or 0.0001% comprising greater than predetermined sequence length.
  • The oligonucleic acids synthesized using the systems and methods described herein are optionally evaluated for sequence accuracy prior to subsequent applications, for example, larger nucleic acid assembly. A common method for oligonucleic acid quality control comprises next generation sequencing.
  • Oligonucleic Acid Release and Assembly
  • Oligonucleic acids synthesized using the methods and substrates described herein, are optionally released from the surface from which they were synthesized. In some cases, oligonucleic acids are cleaved from the surface at this stage. Cleavage may include gas cleavage, e.g., with ammonia or methylamine. In some instances, all the loci in a single cluster collectively correspond to sequence encoding for a single gene, and optionally, when cleaved, remain on the surface of the loci. In some instances, the application of ammonia gas simultaneous deprotects phosphates groups protected during the synthesis steps, i.e., removal of electron-withdrawing cyano group. In some instances, once released from the surface, oligonucleic acids are assembled into larger nucleic acids. Synthesized oligonucleic acids are useful, for example, as components for gene assembly/synthesis, site-directed mutagenesis, nucleic acid amplification, microarrays, and sequencing libraries.
  • In some instances, oligonucleic acids of predetermined sequence are designed to collectively span a large region of a target sequence, such as a gene. In some instances, larger oligonucleic acids are generated through ligation reactions to join the synthesized oligonucleic acids. One example of a ligation reaction is polymerase chain assembly (PCA). In some cases, at least of a portion of the oligonucleic acids are designed to include an appended region that is a substrate for universal primer binding. For PCA reactions, the presynthesized oligonucleic acids include overlaps with each other (e.g., 4, 20, 40 or more bases with overlapping sequence). During the polymerase cycles, the oligonucleic acids anneal to complementary fragments and then are filled in by polymerase. Each cycle thus increases the length of various fragments randomly depending on which oligonucleic acids find each other. Complementarity amongst the fragments allows for forming a complete large span of double stranded DNA. In some cases, after the PCA reaction is complete, an error correction step is conducted using mismatch repair detecting enzymes to remove mismatches in the sequence. Once larger fragments of a target sequence are generated, they can be amplified. For example, in some cases, a target sequence comprising 5′ and 3′ terminal adaptor sequences is amplified in a polymerase chain reaction (PCR) which includes modified primers, e.g., uracil containing primers the hybridize to the adaptor sequences. The use of modified primers allows for removal of the primers through enzymatic reactions centered on targeting the modified base and/or gaps left by enzymes which cleave the modified base pair from the fragment. What remains is a double stranded amplification product that lacks remnants of adapter sequence. In this way, multiple amplification products can be generated in parallel with the same set of primers to generate different fragments of double stranded DNA.
  • In some instances, error correction is performed on synthesized oligonucleic acids and/or assembled products. An example strategy for error correction involves site-directed mutagenesis by overlap extension PCR to correct errors, which is optionally coupled with two or more rounds of cloning and sequencing. In certain instances, double-stranded nucleic acids with mismatches, bulges and small loops, chemically altered bases and/or other heteroduplexes are selectively removed from populations of correctly synthesized nucleic acids by affinity purification. In some instances, error correction is performed using proteins/enzymes that recognize and bind to or next to mismatched or unpaired bases within double stranded nucleic acids to create a single or double strand break or to initiate a strand transfer transposition event. Non-limiting examples of proteins/enzymes for error correction include endonucleases (T7 Endonuclease I, E. coli Endonuclease V, T4 Endonuclease VII, mung bean nuclease, Cell, E. coli Endonuclease IV, UVDE), restriction enzymes, glycosylases, ribonucleases, mismatch repair enzymes, resolvases, helicases, ligases, antibodies specific for mismatches, and their variants. Examples of specific error correction enzymes include T4 endonuclease 7, T7 endonuclease 1, S1, mung bean endonuclease, MutY, MutS, MutH, MutL, cleavase, CELI, and HINF1. In some cases, DNA mismatch-binding protein MutS (Therms aquaticus) is used to remove failure products from a population of synthesized products. In some instances, error correction is performed using the enzyme Correctase. In some cases, error correction is performed using SURVEYOR endonuclease (Transgenomic), a mismatch-specific DNA endonuclease that scans for known and unknown mutations and polymorphisms for heteroduplex DNA.
  • In various instances, a synthesized oligonucleic acid as described herein is amplified in an amplification reaction. In various instances, a nucleic acid assembled from an oligonucleic acid synthesized by the methods and systems described herein is amplified in an amplification reaction. As used herein, at least in some instances, an amplification reaction includes any method known in the art to amplify one or more nucleic acids. Provided herein, in various cases, are instances exemplifying polymerase chain reaction (PCR) as an amplification reaction.
  • In some instances, an amplification reaction, such as PCR, is based on repeated cycles of denaturation, oligonucleic acid primer annealing, and primer extension by thermophilic template dependent polynucleotide polymerase, resulting in the exponential increase in copies of a target nucleic acid sequence flanked by the primers. The two different PCR primers, which anneal to opposite strands of the DNA, are positioned so that the polymerase catalyzed extension product of one primer can serve as a template strand for the other, leading to the accumulation of a discrete double stranded fragment whose length is defined by the distance between the 5′ ends of the oligonucleic acid primers.
  • Systems for Oligonucleic Acid Synthesis
  • Provided herein are systems for the synthesis of oligonucleic acid libraries on a substrate. In some instances, the system comprises the substrate for synthesis support, as described elsewhere herein. In some instances, the system comprises a device for application of one or more reagents of a synthesis method, for example, an oligonucleic acid synthesizer. In some instances, the system comprises a device for treating the substrate with a fluid, for example, a flow cell. In some instances, the system comprises a device for moving the substrate between the application device and the treatment device.
  • In one aspect, provided is an automated system for use with an oligonucleic acid synthesis method described herein that is capable of processing one or more substrates, comprising: a material deposition device for spraying a microdroplet comprising a reagent on a substrate; a scanning transport for scanning the substrate adjacent to the material deposition device to selectively deposit the microdroplet at specified sites; a flow cell for treating the substrate on which the microdroplet is deposited by exposing the substrate to one or more selected fluids; an alignment unit for aligning the substrate correctly relative to the material deposition device each time when the substrate is positioned adjacent to the material deposition device for deposition. In some instances, the system optionally comprises a treating transport for moving the substrate between the material deposition device and the flow cell for treatment in the flow cell, where the treating transport and said scanning transport are different elements. In other instances, the system does not comprise a treating transport.
  • In some instances, a device for application of one or more reagents during a synthesis reagent is an oligonucleic acid synthesizer comprising a plurality of material deposition devices. In some instances, each material deposition device is configured to deposit nucleotide monomers, for example, for phosphoramidite synthesis. In some instances, the oligonucleic acid synthesizer deposits reagents to the resolved loci, wells, and/or microchannels of a substrate. In some cases, the oligonucleic acid synthesizer deposits a drop having a diameter less than about 200 um, 100 um, or 50 um in a volume less than about 1000, 500, 100, 50, or 20 pl. In some cases, the oligonucleic acid synthesizer deposits between about 1 and 10000, 1 and 5000, 100 and 5000, or 1000 and 5000 droplets per second. In some instances, the oligonucleic acid synthesizer uses organic solvents.
  • In some instances, during oligonucleic acid synthesis, the substrate is positioned within or sealed within a flow cell. In some instances, the flow cell provides continuous or discontinuous flow of liquids such as those comprising reagents necessary for reactions within the substrate, for example, oxidizers and/or solvents. In some instances, the flow cell provides continuous or discontinuous flow of a gas, such as nitrogen, for drying the substrate typically through enhanced evaporation of a volatile substrate. A variety of auxiliary devices are useful to improve drying and reduce residual moisture on the surface of the substrate. Examples of such auxiliary drying devices include, without limitation, a vacuum source, depressurizing pump and a vacuum tank. In some cases, an oligonucleic acid synthesis system comprises one or more flow cells, such as 2, 3, 4, 5, 6, 7, 8, 9, 10, or 20 and one or more substrates, such as 2, 3, 4, 5, 6, 7, 8, 9, 10 or 20. In some cases, a flow cell is configured to hold and provide reagents to the substrate during one or more steps in a synthesis reaction. In some instances, a flowcell comprises a lid that slides over the top of a substrate and can be clamped into place to form a pressure tight seal around the edge of the substrate. An adequate seal, includes, without limitation, a seal that allows for about 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 atmospheres of pressure. In some cases, the lid of the flow cell is opened to allow for access to an application device such as an oligonucleic acid synthesizer. In some cases, one or more steps of an oligonucleic acid synthesis method are performed on a substrate within a flow cell, without the transport of the substrate.
  • In some instances, during oligonucleic acid synthesis, a capping element seals with the substrate, to form a resolved reactor. In some instances, a substrate having a plurality of clusters is configured to seal with a capping element having a plurality of caps, wherein when the substrate and capping element are sealed, each cluster is separate from another cluster to form separate resolved reactors for each cluster. In some instances, the capping element is not present in the system or is present and stationary. A resolved reactor is configured to allow for the transfer of fluid, including oligonucleic acids and/or reagents, from the substrate to the capping element and/or vice versa. In some instances, reactors are interconnected or in fluid communication. Fluid communication of reactors allows for washing and perfusion of new reagents for different steps of a synthesis reaction. In some cases, the resolved reactors comprise inlets and/or outlets. In some cases, the inlets and/or outlets are configured for use with a flow cell. As an example, a substrate is sealed within a flow cell where reagents can be introduced and flowed through the substrate, after which the reagents are collected. In some cases, the substrate is drained of fluid and purged with an inert gas such as nitrogen. The flow cell chamber can then be vacuum dried to reduce residual liquids or moisture to less than 1%, 0.1%, 0.01%, 0.001%, 0.0001%, or 0.00001% by volume of the chamber. In some instances, a vacuum chuck is in fluid communication with the substrate for removing gas.
  • In some instances, an oligonucleic acid synthesis system comprises one or more elements useful for downstream processing of the synthesized oligonucleic acids. As an example, the system comprises a temperature control element such as a thermal cycling device. In some instances, the temperature control element is used with a plurality of resolved reactors to perform nucleic acid assembly such as PCA and/or nucleic acid amplification such as PCR.
  • Computer Systems
  • Any of the systems described herein, may be operably linked to a computer and may be automated through a computer either locally or remotely. In various instances, the methods and systems of the invention may further comprise software programs on computer systems and use thereof. Accordingly, computerized control for the synchronization of the dispense/vacuum/refill functions such as orchestrating and synchronizing the material deposition device movement, dispense action and vacuum actuation are within the bounds of the invention. The computer systems may be programmed to interface between the user specified base sequence and the position of a material deposition device to deliver the correct reagents to specified regions of the substrate.
  • The computer system 1300 illustrated in FIG. 13 may be understood as a logical apparatus that can read instructions from media 1311 and/or a network port 1305, which can optionally be connected to server 1309 having fixed media 1312. The system, such as shown in FIG. 13 can include a CPU 1301, disk drives 1303, optional input devices such as keyboard 1315 and/or mouse 1316 and optional monitor 1307. Data communication can be achieved through the indicated communication medium to a server at a local or a remote location. The communication medium can include any means of transmitting and/or receiving data. For example, the communication medium can be a network connection, a wireless connection or an internet connection. Such a connection can provide for communication over the World Wide Web. It is envisioned that data relating to the present disclosure can be transmitted over such networks or connections for reception and/or review by a party 1322 as illustrated in FIG. 13 .
  • FIG. 14 is a block diagram illustrating a first example architecture of a computer system 1400 that can be used in connection with example instances of the present invention. As depicted in FIG. 14 , the example computer system can include a processor 1402 for processing instructions. Non-limiting examples of processors include: Intel Xeon™ processor, AMD Opteron™ processor, Samsung 14-bit RISC ARM 1176JZ(F)-S v1.0™ processor, ARM Cortex-A8 Samsung S5PC100™ processor, ARM Cortex-A8 Apple A4™ processor, Marvell PXA 930™ processor, or a functionally-equivalent processor. Multiple threads of execution can be used for parallel processing. In some instances, multiple processors or processors with multiple cores can also be used, whether in a single computer system, in a cluster, or distributed across systems over a network comprising a plurality of computers, cell phones, and/or personal data assistant devices.
  • As illustrated in FIG. 14 , a high speed cache 1404 can be connected to, or incorporated in, the processor 1402 to provide a high speed memory for instructions or data that have been recently, or are frequently, used by processor 1402. The processor 1402 is connected to a north bridge 1406 by a processor bus 1408. The north bridge 1406 is connected to random access memory (RAM) 1410 by a memory bus 1412 and manages access to the RAM 1410 by the processor 1402. The north bridge 1406 is also connected to a south bridge 1414 by a chipset bus 1416. The south bridge 1414 is, in turn, connected to a peripheral bus 1418. The peripheral bus can be, for example, PCI, PCI-X, PCI Express, or other peripheral bus. The north bridge and south bridge are often referred to as a processor chipset and manage data transfer between the processor, RAM, and peripheral components on the peripheral bus 1418. In some alternative architectures, the functionality of the north bridge can be incorporated into the processor instead of using a separate north bridge chip. In some instances, system 1400 can include an accelerator card 1422 attached to the peripheral bus 1418. The accelerator can include field programmable gate arrays (FPGAs) or other hardware for accelerating certain processing. For example, an accelerator can be used for adaptive data restructuring or to evaluate algebraic expressions used in extended set processing.
  • Software and data are stored in external storage 1424 and can be loaded into RAM 1410 and/or cache 1404 for use by the processor. The system 1400 includes an operating system for managing system resources; non-limiting examples of operating systems include: Linux, Windows™, MACOS™, BlackBerry OS™, iOS™, and other functionally-equivalent operating systems, as well as application software running on top of the operating system for managing data storage and optimization in accordance with example instances of the present invention. In this example, system 1400 also includes network interface cards (NICs) 1420 and 1421 connected to the peripheral bus for providing network interfaces to external storage, such as Network Attached Storage (NAS) and other computer systems that can be used for distributed parallel processing.
  • FIG. 15 is a diagram showing a network 1500 with a plurality of computer systems 1502 a, and 1502 b, a plurality of cell phones and personal data assistants 1502 c, and Network Attached Storage (NAS) 1504 a, and 1504 b. In example instances, systems 1502 a, 1502 b, and 1502 c can manage data storage and optimize data access for data stored in Network Attached Storage (NAS) 1504 a and 1504 b. A mathematical model can be used for the data and be evaluated using distributed parallel processing across computer systems 1502 a, and 1502 b, and cell phone and personal data assistant systems 1502 c. Computer systems 1502 a, and 1502 b, and cell phone and personal data assistant systems 1502 c can also provide parallel processing for adaptive data restructuring of the data stored in Network Attached Storage (NAS) 1504 a and 1504 b. FIG. 15 illustrates an example only, and a wide variety of other computer architectures and systems can be used in conjunction with the various instances of the present invention. For example, a blade server can be used to provide parallel processing. Processor blades can be connected through a back plane to provide parallel processing. Storage can also be connected to the back plane or as Network Attached Storage (NAS) through a separate network interface.
  • In some example instances, processors can maintain separate memory spaces and transmit data through network interfaces, back plane or other connectors for parallel processing by other processors. In other instances, some or all of the processors can use a shared virtual address memory space.
  • FIG. 16 is a block diagram of a multiprocessor computer system 1600 using a shared virtual address memory space in accordance with an example embodiment. The system includes a plurality of processors 1602 a-f that can access a shared memory subsystem 1604. The system incorporates a plurality of programmable hardware memory algorithm processors (MAPs) 1606 a-f in the memory subsystem 1604. Each MAP 1606 a-f can comprise a memory 1608 a-f and one or more field programmable gate arrays (FPGAs) 1610 a-f The MAP provides a configurable functional unit and particular algorithms or portions of algorithms can be provided to the FPGAs 1610 a-f for processing in close coordination with a respective processor. For example, the MAPs can be used to evaluate algebraic expressions regarding the data model and to perform adaptive data restructuring in example instances. In this example, each MAP is globally accessible by all of the processors for these purposes. In one configuration, each MAP can use Direct Memory Access (DMA) to access an associated memory 1608 a-f, allowing it to execute tasks independently of, and asynchronously from the respective microprocessor 1602 a-f. In this configuration, a MAP can feed results directly to another MAP for pipelining and parallel execution of algorithms.
  • The above computer architectures and systems are examples only, and a wide variety of other computer, cell phone, and personal data assistant architectures and systems can be used in connection with example instances, including systems using any combination of general processors, co-processors, FPGAs and other programmable logic devices, system on chips (SOCs), application specific integrated circuits (ASICs), and other processing and logic elements. In some instances, all or part of the computer system can be implemented in software or hardware. Any variety of data storage media can be used in connection with example instances, including random access memory, hard drives, flash memory, tape drives, disk arrays, Network Attached Storage (NAS) and other local or distributed data storage devices and systems.
  • In example instances, the computer system can be implemented using software modules executing on any of the above or other computer architectures and systems. In other instances, the functions of the system can be implemented partially or completely in firmware, programmable logic devices such as field programmable gate arrays (FPGAs) as referenced in FIG. 16 , system on chips (SOCs), application specific integrated circuits (ASICs), or other processing and logic elements. For example, the Set Processor and Optimizer can be implemented with hardware acceleration through the use of a hardware accelerator card, such as accelerator card 1322 illustrated in FIG. 13 .
  • The following examples are set forth to illustrate more clearly the principle and practice of instances disclosed herein to those skilled in the art and are not to be construed as limiting the scope of any claimed instances. Unless otherwise stated, all parts and percentages are on a weight basis.
  • EXAMPLES Example 1: Functionalization of a Substrate Surface
  • A substrate was functionalized to support the attachment and synthesis of a library of oligonucleic acids. The substrate surface was first wet cleaned using a piranha solution comprising 90% H2SO4 and 10% H2O2 for 20 minutes. The substrate was rinsed in several beakers with DI water, held under a DI water gooseneck faucet for 5 min, and dried with N2. The substrate was subsequently soaked in NH4OH (1:100; 3 mL:300 mL) for 5 min, rinsed with DI water using a handgun, soaked in three successive beakers with DI water for 1 min each, and then rinsed again with DI water using the handgun. The substrate was then plasma cleaned by exposing the substrate surface to O2. A SAMCO PC-300 instrument was used to plasma etch O2 at 250 watts for 1 min in downstream mode.
  • The cleaned substrate surface was actively functionalized with a solution comprising N-(3-triethoxysilylpropyl)-4-hydroxybutyramide using a YES-1224P vapor deposition oven system with the following parameters: 0.5 to 1 Torr, 60 min, 70° C., 135° C. vaporizer.
  • The substrate surface was resist coated using a Brewer Science 200× spin coater. SPR™ 3612 photoresist was spin coated on the substrate at 2500 rpm for 40 sec. The substrate was pre-baked for 30 min at 90° C. on a Brewer hot plate. The substrate was subjected to photolithography using a Karl Suss MA6 mask aligner instrument. The substrate was exposed for 2.2 sec and developed for 1 min in MSF 26A. Remaining developer was rinsed with the handgun and the substrate soaked in water for 5 min. The substrate was baked for 30 min at 100° C. in the oven, followed by visual inspection for lithography defects using a Nikon L200. A plasma cleaning process was used to remove residual resist using the SAMCO PC-300 instrument to O2 plasma etch at 250 watts for 1 min.
  • The substrate surface was passively functionalized with a 100 μL solution of perfluorooctyltrichlorosilane mixed with 10 μL light mineral oil. The substrate was placed in a chamber, pumped for 10 min, and then the valve was closed to the pump and left to stand for 10 min. The chamber was vented to air. The substrate was resist stripped by performing two soaks for 5 min in 500 mL NMP at 70° C. with ultrasonication at maximum power (9 on Crest system). The substrate was then soaked for 5 min in 500 mL isopropanol at room temperature with ultrasonication at maximum power. The substrate was dipped in 300 mL of 200 proof ethanol and blown dry with N2. The functionalized surface was activated to serve as a support for oligonucleic acid synthesis.
  • Example 2: Preparation of Substrates Having Distinct Loci Configurations
  • Substrates were manufactured to comprise a plurality of clusters each comprising a plurality of distinct loci configured to provide structural support for oligonucleic acid synthesis. Substrate starting material was a 200 mm standard, double-sided polished silicon wafer having a 725 um thickness. Substrates were processed by a method comprising thermal oxidation at 1000 Å, photolithography using a Karl Suss MA6 mask aligner to generate fiducial structures; oxide etching down to the silicon; and resist stripping. Prepared substrates have 6,144 clusters, with each cluster having 121 reaction sites or loci for oligonucleic acid synthesis. The clusters are organized into 24 sub-fields, which each comprise a 16×16 array of clusters. A schematic of a substrate produced is shown in FIGS. 1-3 . As shown in FIG. 1 , the substrate has a dimension of 140.000 mm by 90.000 mm. As shown in FIG. 2 , the vertical distance between the centers of two adjacent clusters in one substrate is 1079.210 um and in another substrate 1142.694 um. The horizontal distance between the centers of two adjacent clusters in the substrate is 1125.0 um. An expanded view of a cluster of the substrate is shown in FIG. 3 . Each cluster has 121 loci, which are separated so that the horizontal distance between two adjacent loci is 75.000 um and the vertical distance between two loci is 63.483 um. The horizontal distance between the edges of two adjacent loci is 24.0 um.
  • Example 3: Maximization of Microchannel Surface Area
  • Substrates manufactured in this example were processed to generate three-dimensional loci having shapes configured to increase surface area to volume. Examples of locus shapes prepared using the methods described in this example are shown in FIG. 5 . A Silicon on Insulator (SOI) silicon wafer (sub-field size of 32.00×32.00 mm) was oxidized, and the device side processed by photolithography, deep RIE and photoresist stripping. The handle side of the substrate was processed by photolithography, deep RIE, photoresist stripping, and etching by removal of oxide layer (BOX etch). The processed substrate has a plurality of wells or holes within the handle layer, each having a width of 1.150 mm, wherein each channel has a plurality of microchannels having shapes that allow for an increase in surface area to volume. The smallest etch size for a feature of a shape of a microchannel within a substrate prepared in this example was 5 um. The distance between the centers of two adjacent clusters (wells) is 1.693 mm in all directions. The distance between the centers of two adjacent loci (microchannels) is 97.765 in a horizontal direction and 84.667 um in a vertical direction. The prepared substrate has a set of markings or fiducials of 0.5 mm diameter. The width of the main channel is 1.150 mm and the width of the microchannel is 5 um. Detailed features of substrates prepared using these methods are shown in FIGS. 5-8 .
  • A cluster of a processed substrate having double comb shaped loci is shown in a bird's eye view in FIG. 6 . The combined height of the two longest teeth is 57 um. The distance between two teeth of the comb is 14.0 um. The width of the comb handle is 5 um. The combined height of the two shortest teeth is 38.0 um. The width of the comb in the horizontal direction is 47.0 urn.
  • A cluster of a processed substrate having single comb shaped loci is shown in a bird's eye view in FIG. 7 . The height of the longest tooth is 49.0 um. The distance between two teeth of the comb is 14.0 um. The width of the comb handle is 5 um. The height of the shortest tooth is 39.0 um. The width of the comb in the horizontal direction is 47.0 um.
  • A cluster of a processed substrate having serpentine shaped loci is shown in a bird's eye view in FIG. 8 . The height of the loci shape is 54 um. The distance between two lines of the shape is 14 um. The width of a line of the shape is 5 um.
  • Detailed measurements for prepared substrates are shown in Table 3.
  • TABLE 3
    Device depth (um) 30
    Width of segments (um) 5
    Top surface Total
    Internal growth projected Total Total
    area area Volume area Area Volume
    Segments No. (um2) (um2) (um3) (um2) (um2) (um3)
    Double comb
    Lateral segments
    3 540 45 1350 135 1620 4050
    End segments 2 1890 170 5100 340 3780 10200
    Middle segments 2 2850 263 7875 525 5700 15750
    Rounded ends 4 471 20 589 79 1885 2356
    Total 1079 12985 32356
    Total growth area 15225
    including top surface (um2)
    Single comb
    Lateral segments 1 2456 210 7050 210 2456 7050
    End segments 2 2070 173 5175 345 4140 10350
    Middle segments 2 2490 208 6225 415 4980 12450
    Rounded ends 2 471 20 589 39 942 1178
    Total 1009 12518 31028
    Total growth area 14827
    including top surface (um2)
    Serpentine
    Vertical segments
    4 2100 175 5250 700 8400 21000
    Annulus segments (whole) 1.5 2639 220 6597 330 3958 9896
    Rounded ends 1 471 20 589 20 471 589
    Total 1050 12830 31485
    Total growth area 15098
    including top surface (um2)
  • The surface and volume parameters of loci having high surface area shapes (double comb, single comb, and serpentine) prepared using these methods were compared with the parameters of a locus having a revolver shape (barrel comprising 5 channels). The comparison is shown in Table 4. The loci having a comb or serpentine shape had a lower substrate volume than a substrate having revolver loci. The loci having a comb or serpentine shape had a greater surface area than a substrate having revolver loci. The loci having a comb or serpentine shape had a greater surface area to volume ratio than a substrate having revolver loci.
  • TABLE 4
    Revolver Double comb Single come Serpentine
    Total volume 47 32 31 31
    (pL)
    Total surface 9425 12985 12518 12830
    area (um)
    Surface area to 0.20 0.40 0.40 0.41
    volume ratio
    (1/um)
    Total volume 1.00 0.69 0.66 0.67
    relative to
    revolver
    Total surface 1.00 1.38 1.33 1.36
    area relative to
    revolver
    Surface area to 1.00 2.01 2.02 2.04
    volume ratio
    relative revolver
  • Example 4: Synthesis of a 100-Mer Oligonucleic Acid on a Substantially Planar Substrate
  • A substantially planar substrate functionalized for oligonucleic acid synthesis was assembled into a flow cell and connected to an Applied Biosystems ABI394 DNA Synthesizer. In one experiment, the substrate was uniformly functionalized with N-(3-triethoxysilylpropyl)-4-hydroxybutyramide. In another experiment, the substrate was functionalized with a 5/95 mix of 11-acetoxyundecyltriethoxysilane and N-decyltriethoxysilane. Synthesis of 100-mer oligonucleic acids (“100-mer oligonucleotide”; 5′: CGGGATCCTTATCGTCATCGTCGTACAGATCCCGACCCATTTGCTGTCCACCAGT CATGCTAGCCATACCATGATGATGATGATGATGAGAACCCCGCAT##TTTTTTTTT T 3′ (SEQ ID NO: 1), where #denotes Thymidine-succinyl hexamide CED phosphoramidite (CLP-2244 from ChemGenes)) were performed using the methods of Table 5.
  • TABLE 5
    General DNA Synthesis Time
    Process Name Process Step (sec)
    WASH (Acetonitrile Wash Acetonitrile System Flush 4
    Flow) Acetonitrile to Flowcell 23
    N2 System Flush 4
    Acetonitrile System Flush 4
    DNA BASE ADDITION Activator Manifold Flush 2
    (Phosphoramidite + Activator to Flowcell 6
    Activator Flow) Activator + 6
    Phosphoramidite to
    Flowcell
    Activator to Flowcell 0.5
    Activator + 5
    Phosphoramidite to
    Flowcell
    Activator to Flowcell 0.5
    Activator + 5
    Phosphoramidite to
    Flowcell
    Activator to Flowcell 0.5
    Activator + 5
    Phosphoramidite to
    Flowcell
    Incubate for 25 sec 25
    WASH (Acetonitrile Wash Acetonitrile System Flush 4
    Flow) Acetonitrile to Flowcell 15
    N2 System Flush 4
    Acetonitrile System Flush 4
    DNA BASE ADDITION Activator Manifold Flush 2
    (Phosphoramidite + Activator to Flowcell 5
    Activator Flow) Activator + 18
    Phosphoramidite to
    Flowcell
    Incubate for 25 sec 25
    WASH (Acetonitrile Wash Acetonitrile System Flush 4
    Flow) Acetonitrile to Flowcell 15
    N2 System Flush 4
    Acetonitrile System Flush 4
    CAPPING (CapA + B, 1:1, CapA + B to Flowcell 15
    Flow)
    WASH (Acetonitrile Wash Acetonitrile System Flush 4
    Flow) Acetonitrile to Flowcell 15
    Acetonitrile System Flush 4
    OXIDATION (Oxidizer Oxidizer to Flowcell 18
    Flow)
    WASH (Acetonitrile Wash Acetonitrile System Flush 4
    Flow) N2 System Flush 4
    Acetonitrile System Flush 4
    Acetonitrile to Flowcell 15
    Acetonitrile System Flush 4
    Acetonitrile to Flowcell 15
    N2 System Flush 4
    Acetonitrile System Flush 4
    Acetonitrile to Flowcell 23
    N2 System Flush 4
    Acetonitrile System Flush 4
    DEBLOCKING (Deblock Deblock to Flowcell 36
    Flow)
    WASH (Acetonitrile Wash Acetonitrile System Flush 4
    Flow) N2 System Flush 4
    Acetonitrile System Flush 4
    Acetonitrile to Flowcell 18
    N2 System Flush 4.13
    Acetonitrile System Flush 4.13
    Acetonitrile to Flowcell 15
  • Synthesized oligonucleic acids were extracted from the substrate surface and analyzed on a BioAnalyzer chip. Oligonucleic acid products were PCR amplified, cloned and Sanger sequenced. Table 6 summarizes the Sanger sequencing results for samples taken from spots 1-5 from one chip and spots 6-10 from a second chip.
  • TABLE 6
    Spot Error rate Cycle efficiency
    1  1/763 bp 99.87%
    2  1/824 bp 99.88%
    3  1/780 bp 99.87%
    4  1/429 bp 99.77%
    5 1/1525 bp 99.93%
    6 1/1615 bp 99.94%
    7  1/531 bp 99.81%
    8 1/1769 bp 99.94%
    9  1/854 bp 99.88%
    10 1/1451 bp 99.93%
  • Overall, 89% (233/262) of the 100-mers that were sequenced had sequences without errors. Table 7 summarizes key error characteristics for the sequences obtained from the oligonucleic acid samples from spots 1-10.
  • TABLE 7
    Sample ID/ OSA_ OSA_ OSA_ OSA_ OSA_ OSA_ OSA_ OSA_ OSA_ OSA_
    Spot No. 0046/1 0047/2 0048/3 0049/4 0050/5 0051/6 0052/7 0053/8 0054/9 0055/10
    Total 32 32 32 32 32 32 32 32 32 32
    Sequences
    Sequencing 25 of 27 of 26 of 21 of 25 of 29 of 27 of 29 of 28 of 25 of
    Quality 28 27 30 23 26 30 31 31 29 28
    Oligo 23 of 25 of 22 of 18 of 24 of 25 of 22 of 28 of 26 of 20 of
    Quality 25 27 26 21 25 29 27 29 28 25
    ROI Match 2500 2698 2561 2122 2499 2666 2625 2899 2798 2348
    Count
    ROI 2 2 1 3 1 0 2 1 2 1
    Mutation
    ROI Multi 0 0 0 0 0 0 0 0 0 0
    Base
    Deletion
    ROI Small 1 0 0 0 0 0 0 0 0 0
    Insertion
    ROI Single 0 0 0 0 0 0 0 0 0 0
    Base
    Deletion
    Large 0 0 1 0 0 1 1 0 0 0
    Deletion
    Count
    Mutation: 2 2 1 2 1 0 2 1 2 1
    G > A
    Mutation: 0 0 0 1 0 0 0 0 0 0
    T > C
    ROI Error 3 2 2 3 1 1 3 1 2 1
    Count
    ROI Error Err:~1 Err:~1 Err:~1 Err:~1 Err:~1 Err:~1 Err:~1 Err:~1 Err:~1 Err:~1
    Rate in 834 in 1350 in 1282 in 708 in 2500 in 2667 in 876 in 2900 in 1400 in 2349
    ROI Minus MP MP MP MP MP MP MP MP MP MP
    Primer Err:~1 Err:~1 Err:~1 Err:~1 Err:~1 Err:~1 Err:~1 Err:~1 Err:~1 Err:~1
    Error Rate in 763 in 824 in 780 in 429 in 1525 in 1615 in 531 in 1769 in 854 in 1451
  • Example 5: Gene Assembly in Reactors Using PCA
  • Gene assembly within nanoreactors created using a three-dimensional substrate was performed. PCA reactions were performed using oligonucleic acids described in Table 8 (SEQ ID NOS: 2-61) to assemble the 3075 base LacZ gene (SEQ ID NO.: 62) using the reaction mixture of Table 9 within individual nanoreactors.
  • TABLE 8
    Sequence Name Sequence
    Oligo_1, 5′ATGACCATGATTACGGATTCACTGGCCG
    SEQ ID NO.: 2 TCGTTTTACAACGTCGTGACTGGGAAAACC
    CTGG3′
    Oligo_2, 5′GCCAGCTGGCGAAAGGGGGATGTGCTGC
    SEQ ID NO.: 3 AAGGCGATTAAGTTGGGTAACGCCAGGGTT
    TTCCCAGTCACGAC3′
    Oligo_3, 5′CCCCCTTTCGCCAGCTGGCGTAATAGCG
    SEQ ID NO.: 4 AAGAGGCCCGCACCGATCGCCCTTCCCAAC
    AGTTGCGCAGCC3′
    Oligo_4, 5′CGGCACCGCTTCTGGTGCCGGAAACCAG
    SEQ ID NO.: 5 GCAAAGCGCCATTCGCCATTCAGGCTGCGC
    AACTGTTGGGA3′
    Oligo_5, 5′CACCAGAAGCGGTGCCGGAAAGCTGGCT
    SEQ ID NO.: 6 GGAGTGCGATCTTCCTGAGGCCGATACTGT
    CGTCGTCCCCTC3′
    Oligo_6, 5′GATAGGTCACGTTGGTGTAGATGGGCGC
    SEQ ID NO.: 7 ATCGTAACCGTGCATCTGCCAGTTTGAGGG
    GACGACGACAGTATCGG3′
    Oligo_7, 5′CCCATCTACACCAACGTGACCTATCCCA
    SEQ ID NO.: 8 TTACGGTCAATCCGCCGTTTGTTCCCACGG
    AGAATCCGACGGGTTG3′
    Oligo_8, 5′GTCTGGCCTTCCTGTAGCCAGCTTTCAT
    SEQ ID NO.: 9 CAACATTAAATGTGAGCGAGTAACAACCCG
    TCGGATTCTCCGTG3′
    Oligo_9, 5′GCTGGCTACAGGAAGGCCAGACGCGAAT
    SEQ ID NO.: 10 TATTTTTGATGGCGTTAACTCGGCGTTTCA
    TCTGTGGTGCAACGG3′
    Oligo_10, 5′CAGGTCAAATTCAGACGGCAAACGACTG
    SEQ ID NO.: 11 TCCTGGCCGTAACCGACCCAGCGCCCGTTG
    CACCACAGATGAAACG3′
    Oligo_11, 5′CGTTTGCCGTCTGAATTTGACCTGAGCG
    SEQ ID NO.: 12 CATTTTTACGCGCCGGAGAAAACCGCCTCG
    CGGTGATGGTGCTG3′
    Oligo_12, 5′GCCGCTCATCCGCCACATATCCTGATCT
    SEQ ID NO.: 13 TCCAGATAACTGCCGTCACTCCAGCGCAGC
    ACCATCACCGCGAG3′
    Oligo_13, 5′AGGATATGTGGCGGATGAGCGGCATTTT
    SEQ ID NO.: 14 CCGTGACGTCTCGTTGCTGCATAAACCGAC
    TACACAAATCAGCGATTTC3′
    Oligo_14, 5′CTCCAGTACAGCGCGGCTGAAATCATCA
    SEQ ID NO.: 15 TTAAAGCGAGTGGCAACATGGAAATCGCTG
    ATTTGTGTAGTCGGTTTATG3′
    Oligo_15, 5′ATTTCAGCCGCGCTGTACTGGAGGCTGA
    SEQ ID NO.: 16 AGTTCAGATGTGCGGCGAGTTGCGTGACTA
    CCTACGGGTAACAGTTT3
    Oligo_16, 5′AAAGGCGCGGTGCCGCTGGCGACCTGCG
    SEQ ID NO.: 17 TTTCACCCTGCCATAAAGAAACTGTTACCC
    GTAGGTAGTCACG3′
    Oligo_17, 5′GCGGCACCGCGCCTTTCGGCGGTGAAAT
    SEQ ID NO.: 18 TATCGATGAGCGTGGTGGTTATGCCGATCG
    CGTCACACTACG3′
    Oligo_18, 5′GATAGAGATTCGGGATTTCGGCGCTCCA
    SEQ ID NO.: 19 CAGTTTCGGGTTTTCGACGTTCAGACGTAG
    TGTGACGCGATCGGCA3′
    Oligo_19, 5′GAGCGCCGAAATCCCGAATCTCTATCGT
    SEQ ID NO.: 20 GCGGTGGTTGAACTGCACACCGCCGACGGC
    ACGCTGATTGAAGCAG3′
    Oligo_20, 5′CAGCAGCAGACCATTTTCAATCCGCACC
    SEQ ID NO.: 21 TCGCGGAAACCGACATCGCAGGCTTCTGCT
    TCAATCAGCGTGCCG3′
    Oligo_21, 5′CGGATTGAAAATGGTCTGCTGCTGCTGA
    SEQ ID NO.: 22 ACGGCAAGCCGTTGCTGATTCGAGGCGTTA
    ACCGTCACGAGCATCA3′
    Oligo_22, 5′GCAGGATATCCTGCACCATCGTCTGCTC
    SEQ ID NO.: 23 ATCCATGACCTGACCATGCAGAGGATGATG
    CTCGTGACGGTTAACGC3′
    Oligo_23, 5′CAGACGATGGTGCAGGATATCCTGCTGA
    SEQ ID NO.: 24 TGAAGCAGAACAACTTTAACGCCGTGCGCT
    GTTCGCATTATCCGAAC3′
    Oligo_24, 5′TCCACCACATACAGGCCGTAGCGGTCGC
    SEQ ID NO.: 25 ACAGCGTGTACCACAGCGGATGGTTCGGAT
    AATGCGAACAGCGCAC3′
    Oligo25, 5′GCTACGGCCTGTATGTGGTGGATGAAGC
    SEQ ID NO.:  CAATATTGAAACCCACGGCATGGTGCCAAT
    26 GAATCGTCTGACCGATG3′
    Oligo_26, 5′GCACCATTCGCGTTACGCGTTCGCTCAT
    SEQ ID NO.: 27 CGCCGGTAGCCAGCGCGGATCATCGGTCAG
    ACGATTCATTGGCAC3′
    Oligo_27, 5′CGCGTAACGCGAATGGTGCAGCGCGATC
    SEQ ID NO.: 28 GTAATCACCCGAGTGTGATCATCTGGTCGC
    TGGGGAATGAATCAG3′
    Oligo_28, 5′GGATCGACAGATTTGATCCAGCGATACA
    SEQ ID NO.: 29 GCGCGTCGTGATTAGCGCCGTGGCCTGATT
    CATTCCCCAGCGACCAGATG3′
    Oligo_29, 5′GTATCGCTGGATCAAATCTGTCGATCCT
    SEQ ID NO.: 30 TCCCGCCCGGTGCAGTATGAAGGCGGCGGA
    GCCGACACCACGGC3′
    Oligo_30, 5′CGGGAAGGGCTGGTCTTCATCCACGCGC
    SEQ ID NO.: 31 GCGTACATCGGGCAAATAATATCGGTGGCC
    GTGGTGTCGGCTC3′
    Oligo_31, 5′TGGATGAAGACCAGCCCTTCCCGGCTGT
    SEQ ID NO.: 32 GCCGAAATGGTCCATCAAAAAATGGCTTTC
    GCTACCTGGAGAGAC3′
    Oligo_32, 5′CCAAGACTGTTACCCATCGCGTGGGCGT
    SEQ ID NO.: 33 ATTCGCAAAGGATCAGCGGGCGCGTCTCTC
    CAGGTAGCGAAAGCC3′
    Oligo_33, 5′CGCGATGGGTAACAGTCTTGGCGGTTTC
    SEQ ID NO.: 34 GCTAAATACTGGCAGGCGTTTCGTCAGTAT
    CCCCGTTTACAGGGC3′
    Oligo_34, 5′GCCGTTTTCATCATATTTAATCAGCGAC
    SEQ ID NO.: 35 TGATCCACCCAGTCCCAGACGAAGCCGCCC
    TGTAAACGGGGATACTGACG3′
    Oligo_35, 5′CAGTCGCTGATTAAATATGATGAAAACG
    SEQ ID NO.: 36 GCAACCCGTGGTCGGCTTACGGCGGTGATT
    TTGGCGATACGCCGAACG3′
    Oligo_36, 5′GCGGCGTGCGGTCGGCAAAGACCAGACC
    SEQ ID NO.: 31 GTTCATACAGAACTGGCGATCGTTCGGCGT
    ATCGCCAAA3′
    Oligo_37, 5′CGACCGCACGCCGCATCCAGCGCTGACG
    SEQ ID NO.: 38 GAAGCAAAACACCAGCAGCAGTTTTTCCAG
    TTCCGTTTATCCG3′
    Oligo_38, 5′CTCGTTATCGCTATGACGGAACAGGTAT
    SEQ ID NO.: 39 TCGCTGGTCACTTCGATGGTTTGCCCGGAT
    AAACGGAACTGGAAAAACTGC3′
    Oligo_39, 5′AATACCTGTTCCGTCATAGCGATAACGA
    SEQ ID NO.: 40 GCTCCTGCACTGGATGGTGGCGCTGGATGG
    TAAGCCGCTGGCAAGCG3′
    Oligo_40, 5′GTTCAGGCAGTTCAATCAACTGTTTACC
    SEQ ID NO.: 41 TTGTGGAGCGACATCCAGAGGCACTTCACC
    GCTTGCCAGCGGCTTACC3’
    Oligo_41, 5′CAAGGTAAACAGTTGATTGAACTGCCTG
    SEQ ID NO.: 42 AACTACCGCAGCCGGAGAGCGCCGGGCAAC
    TCTGGCTCACAGTACGCGTA3′
    Oligo_42, 5′GCGCTGATGTGCCCGGCTTCTGACCATG
    SEQ ID NO.: 43 CGGTCGCGTTCGGTTGCACTACGCGTACTG
    TGAGCCAGAGTTG3′
    Oligo_43, 5′CCGGGCACATCAGCGCCTGGCAGCAGTG
    SEQ ID NO.: 44 GCGTCTGGCGGAAAACCTCAGTGTGACGCT
    CCCCGCCGC3′
    Oligo_44, 5′CCAGCTCGATGCAAAAATCCATTTCGCT
    SEQ ID NO.: 45 GGTGGTCAGATGCGGGATGGCGTGGGACGC
    GGCGGGGAGCGTC3′
    Oligo_45, 5′CGAAATGGATTTTTGCATCGAGCTGGGT
    SEQ ID NO.: 46 AATAAGCGTTGGCAATTTAACCGCCAGTCA
    GGCTTTCTTTCACAGATGTG3′
    Oligo_46, 5′TGAACTGATCGCGCAGCGGCGTCAGCAG
    SEQ ID NO.: 47 TTGTTTTTTATCGCCAATCCACATCTGTGA
    AAGAAAGCCTGACTGG3′
    Oligo_47, 5′GCCGCTGCGCGATCAGTTCACCCGTGCA
    SEQ ID NO.: 48 CCGCTGGATAACGACATTGGCGTAAGTGAA
    GCGACCCGCATTGAC3′
    Oligo_48, 5′GGCCTGGTAATGGCCCGCCGCCTTCCAG
    SEQ ID NO.: 49 CGTTCGACCCAGGCGTTAGGGTCAATGCGG
    GTCGCTTCACTTA3′
    Oligo_49, 5′CGGGCCATTACCAGGCCGAAGCAGCGTT
    SEQ ID NO.: 50 GTTGCAGTGCACGGCAGATACACTTGCTGA
    TGCGGTGCTGAT3′
    Oligo_50, 5′TCCGGCTGATAAATAAGGTTTTCCCCTG
    SEQ ID NO.: 51 ATGCTGCCACGCGTGAGCGGTCGTAATCAG
    CACCGCATCAGCAAGTG3′
    Oligo_51, 5′GGGGAAAACCTTATTTATCAGCCGGAAA
    SEQ ID NO.: 52 ACCTACCGGATTGATGGTAGTGGTCAAATG
    GCGATTACCGTTGATGTTGA3′
    Oligo_52, 5′GGCAGTTCAGGCCAATCCGCGCCGGATG
    SEQ ID NO.: 53 CGGTGTATCGCTCGCCACTTCAACATCAAC
    GGTAATCGCCATTTGAC3′
    Oligo_53, 5′GCGGATTGGCCTGAACTGCCAGCTGGCG
    SEQ ID NO.: 54 CAGGTAGCAGAGCGGGTAAACTGGCTCGGA
    TTAGGGCCGCAAG3′
    Oligo_54, 5′GGCAGATCCCAGCGGTCAAAACAGGCGG
    SEQ ID NO.: 55 CAGTAAGGCGGTCGGGATAGTTTTCTTGCG
    GCCCTAATCCGAGC3′
    Oligo55, 5′GTTTTGACCGCTGGGATCTGCCATTGTC
    SEQ ID NO.: 56 AGACATGTATACCCCGTACGTCTTCCCGAG
    CGAAAACGGTCTGC3′
    Oligo_56, 5′GTCGCCGCGCCACTGGTGTGGGCCATAA
    SEQ ID NO.: 57 TTCAATTCGCGCGTCCCGCAGCGCAGACCG
    TTTTCGCTCGG3′
    Oligo_57, 5′ACCAGTGGCGCGGCGACTTCCAGTTCAA
    SEQ ID NO.: 58 CATCAGCCGCTACAGTCAACAGCAACTGAT
    GGAAACCAGCCATC3′
    Oligo_58, 5′GAAACCGTCGATATTCAGCCATGTGCCT
    SEQ ID NO.: 59 TCTTCCGCGTGCAGCAGATGGCGATGGCTG
    GTTTCCATCAGTTGCTG3′
    Oligo_59, 5′CATGGCTGAATATCGACGGTTTCCATAT
    SEQ ID NO.: 60 GGGGATTGGTGGCGACGACTCCTGGAGCCC
    GTCAGTATCGGCG3′
    Oligo_60, 5′TTATTTTTGACACCAGACCAACTGGTAA
    SEQ ID NO.: 61 TGGTAGCGACCGGCGCTCAGCTGGAATTCC
    GCCGATACTGACGGGC3′
    LacZ gene- 5′ATGACCATGATTACGGATTCACTGGCCG
    SEQ ID NO: 62 TCGTTTTACAACGTCGTGACTGGGAAAACC
    CTGGCGTTACCCAACTTAATCGCCTTGCAG
    CACATCCCCCTTTCGCCAGCTGGCGTAATA
    GCGAAGAGGCCCGCACCGATCGCCCTTCCC
    AACAGTTGCGCAGCCTGAATGGCGAATGGC
    GCTTTGCCTGGTTTCCGGCACCAGAAGCGG
    TGCCGGAAAGCTGGCTGGAGTGCGATCTTC
    CTGAGGCCGATACTGTCGTCGTCCCCTCAA
    ACTGGCAGATGCACGGTTACGATGCGCCCA
    TCTACACCAACGTGACCTATCCCATTACGG
    TCAATCCGCCGTTTGTTCCCACGGAGAATC
    CGACGGGTTGTTACTCGCTCACATTTAATG
    TTGATGAAAGCTGGCTACAGGAAGGCCAGA
    CGCGAATTATTTTTGATGGCGTTAACTCGG
    CGTTTCATCTGTGGTGCAACGGGCGCTGGG
    TCGGTTACGGCCAGGACAGTCGTTTGCCGT
    CTGAATTTGACCTGAGCGCATTTTTACGCG
    CCGGAGAAAACCGCCTCGCGGTGATGGTGC
    TGCGCTGGAGTGACGGCAGTTATCTGGAAG
    ATCAGGATATGTGGCGGATGAGCGGCATTT
    TCCGTGACGTCTCGTTGCTGCATAAACCGA
    CTACACAAATCAGCGATTTCCATGTTGCCA
    CTCGCTTTAATGATGATTTCAGCCGCGCTG
    TACTGGAGGCTGAAGTTCAGATGTGCGGCG
    AGTTGCGTGACTACCTACGGGTAACAGTTT
    CTTTATGGCAGGGTGAAACGCAGGTCGCCA
    GCGGCACCGCGCCTTTCGGCGGTGAAATTA
    TCGATGAGCGTGGTGGTTATGCCGATCGCG
    TCACACTACGTCTGAACGTCGAAAACCCGA
    AACTGTGGAGCGCCGAAATCCCGAATCTCT
    ATCGTGCGGTGGTTGAACTGCACACCGCCG
    ACGGCACGCTGATTGAAGCAGAAGCCTGCG
    ATGTCGGTTTCCGCGAGGTGCGGATTGAAA
    ATGGTCTGCTGCTGCTGAACGGCAAGCCGT
    TGCTGATTCGAGGCGTTAACCGTCACGAGC
    ATCATCCTCTGCATGGTCAGGTCATGGATG
    AGCAGACGATGGTGCAGGATATCCTGCTGA
    TGAAGCAGAACAACTTTAACGCCGTGCGCT
    GTTCGCATTATCCGAACCATCCGCTGTGGT
    ACACGCTGTGCGACCGCTACGGCCTGTATG
    TGGTGGATGAAGCCAATATTGAAACCCACG
    GCATGGTGCCAATGAATCGTCTGACCGATG
    ATCCGCGCTGGCTACCGGCGATGAGCGAAC
    GCGTAACGCGAATGGTGCAGCGCGATCGTA
    ATCACCCGAGTGTGATCATCTGGTCGCTGG
    GGAATGAATCAGGCCACGGCGCTAATCACG
    ACGCGCTGTATCGCTGGATCAAATCTGTCG
    ATCCTTCCCGCCCGGTGCAGTATGAAGGCG
    GCGGAGCCGACACCACGGCCACCGATATTA
    TTTGCCCGATGTACGCGCGCGTGGATGAAG
    ACCAGCCCTTCCCGGCTGTGCCGAAATGGT
    CCATCAAAAAATGGCTTTCGCTACCTGGAG
    AGACGCGCCCGCTGATCCTTTGCGAATACG
    CCCACGCGATGGGTAACAGTCTTGGCGGTT
    TCGCTAAATACTGGCAGGCGTTTCGTCAGT
    ATCCCCGTTTACAGGGCGGCTTCGTCTGGG
    ACTGGGTGGATCAGTCGCTGATTAAATATG
    ATGAAAACGGCAACCCGTGGTCGGCTTACG
    GCGGTGATTTTGGCGATACGCCGAACGATC
    GCCAGTTCTGTATGAACGGTCTGGTCTTTG
    CCGACCGCACGCCGCATCCAGCGCTGACGG
    AAGCAAAACACCAGCAGCAGTTTTTCCAGT
    TCCGTTTATCCGGGCAAACCATCGAAGTGA
    CCAGCGAATACCTGTTCCGTCATAGCGATA
    ACGAGCTCCTGCACTGGATGGTGGCGCTGG
    ATGGTAAGCCGCTGGCAAGCGGTGAAGTGC
    CTCTGGATGTCGCTCCACAAGGTAAACAGT
    TGATTGAACTGCCTGAACTACCGCAGCCGG
    AGAGCGCCGGGCAACTCTGGCTCACAGTAC
    GCGTAGTGCAACCGAACGCGACCGCATGGT
    CAGAAGCCGGGCACATCAGCGCCTGGCAGC
    AGTGGCGTCTGGCGGAAAACCTCAGTGTGA
    CGCTCCCCGCCGCGTCCCACGCCATCCCGC
    ATCTGACCACCAGCGAAATGGATTTTTGCA
    TCGAGCTGGGTAATAAGCGTTGGCAATTTA
    ACCGCCAGTCAGGCTTTCTTTCACAGATGT
    GGATTGGCGATAAAAAACAACTGCTGACGC
    CGCTGCGCGATCAGTTCACCCGTGCACCGC
    TGGATAACGACATTGGCGTAAGTGAAGCGA
    CCCGCATTGACCCTAACGCCTGGGTCGAAC
    GCTGGAAGGCGGCGGGCCATTACCAGGCCG
    AAGCAGCGTTGTTGCAGTGCACGGCAGATA
    CACTTGCTGATGCGGTGCTGATTACGACCG
    CTCACGCGTGGCAGCATCAGGGGAAAACCT
    TATTTATCAGCCGGAAAACCTACCGGATTG
    ATGGTAGTGGTCAAATGGCGATTACCGTTG
    ATGTTGAAGTGGCGAGCGATACACCGCATC
    CGGCGCGGATTGGCCTGAACTGCCAGCTGG
    CGCAGGTAGCAGAGCGGGTAAACTGGCTCG
    GATTAGGGCCGCAAGAAAACTATCCCGACC
    GCCTTACTGCCGCCTGTTTTGACCGCTGGG
    ATCTGCCATTGTCAGACATGTATACCCCGT
    ACGTCTTCCCGAGCGAAAACGGTCTGCGCT
    GCGGGACGCGCGAATTGAATTATGGCCCAC
    ACCAGTGGCGCGGCGACTTCCAGTTCAACA
    TCAGCCGCTACAGTCAACAGCAACTGATGG
    AAACCAGCCATCGCCATCTGCTGCACGCGG
    AAGAAGGCACATGGCTGAATATCGACGGTT
    TCCATATGGGGATTGGTGGCGACGACTCCT
    GGAGCCCGTCAGTATCGGCGGAATTCCAGC
    TGAGCGCCGGTCGCTACCATTACCAGTTGG
    TCTGGTGTCAAAAATAA3′
  • TABLE 9
    PCA reaction mixture 1 (×100 ul) Final conc.
    H2O 62.00
    5x Q5 buffer 20.00 1x
    10 mM dNTP 1.00 100 uM
    BSA
    20 mg/ml 5.00  1 mg/ml
    Oligonucleic acid mix (50 nM each) 10.00  5 nM
    Q5 polymerase 2U/ul 2.00 2 U/50 ul
  • PCA reaction mixture drops of about 400 nL were dispensed using a Mantis dispenser (Formulatrix, MA) on the top of channels of a device side of a three-dimensional substrate having a plurality of loci microchannels in fluid communication with a single main channel of a cluster. A nanoreactor chip was manually mated with the substrate to pick up the droplets having the PCA reaction mixture and oligonucleic acids from each channel. The droplets were picked up into individual nanoreactors in the nanoreactor chip by releasing the nanoreactor from the substrate immediately after pick up. The nanoreactors were sealed with a heat sealing film, placed in a thermocycler for PCA. PCA thermocycling conditions are shown in Table 10. An aliquot of 0.5 ul was collected from 1-10 individual wells and the aliquots were amplified in plastic PCR tubes using forward primer (5′ATGACCATGATTACGGATTCACTGGCC3′ SEQ ID NO.: 63) and reverse primer (5′TTATTTTTGACACCAGACCAACTGGTAATGG3′ SEQ ID NO.: 64). Thermocycling conditions for PCR are shown in Table 11 and PCR reaction components are shown in Table 12. The amplification products were ran on a BioAnalyzer instrument and on a gel. The gel showed products 1-10 having a size slightly larger than 3000 bp (data not shown). A PCA reaction performed in plastic tube was also ran on the gel as a positive control (panel 11), which shows a product having a similar size to the products from wells 1-10. A negative control (panel 12) was also run on the gel, which corresponds to a PCR reaction ran without a PCA template. The BioAnalyzer data is not shown.
  • TABLE 10
    No. of cycles Temperature (° C.) Time
    1 98 45 seconds
    40 98 15 seconds
    63 45 seconds
    72 60 seconds
    1 72  5 minutes
    1 4 Hold
  • TABLE 11
    No. of cycles Temperature (° C.) Time
    1 98 30 seconds
    30 98  7 seconds
    63 30 seconds
    72 90 seconds
    1 72  5 minutes
    1 4 Hold
  • TABLE 12
    PCR 1 (×25 ul) Final conc.
    H2O 17.50
    5x Q5 buffer 5.00 1x
    10 mM dNTP 0.50  200 uM
    F-primer 20 uM 0.63  0.5 uM
    R-primer 20 uM 0.63  0.5 uM
    BSA
    20 mg/ml 0.00
    Q5 pol 2U/ul 0.25 1U/50 ul
    Template (PCA assembly) 0.50 1 ul/50 ul rxn
  • Example 6: Error Correction of Assembled Nucleic Acids
  • A gene of about 1 kbp (SEQ ID.: 67; Table 13) was assembled using 6 purchased Ultramer oligonucleotides (SEQ ID NO.: 68-73; Table 13) and assembled in a PCA reaction. Ultramer oligonucleotides are expected to have error rates of at least 1 in 500 to 1 in 200 nucleotides. The assembled gene was amplified by PCR using a forward primer (5′ ATGACCATGATTACGGATTCACTGGCC3′ SEQ ID NO.: 65) and a reverse primer (5′GATAGAGATTCGGGATTTCGGCGCTCC3′ SEQ ID NO.: 66). The amplified assembled gene was analyzed in a BioAnalyzer and cloned. DNA preparations from 24 colonies were Sanger sequenced. The BioAnalyzer analysis provided a broad peak and a tail for the uncorrected gene, indicating a high error rate. The sequencing indicated an error rate of 1/789 bases. Two rounds of error correction were followed using CorectASE (Life Technologies) according to the manufacturer's instructions. The resulting gene samples were analyzed in a BioAnalyzer after round one and round two and cloned. Twenty-four colonies were picked for sequencing. The sequencing results indicated an error rate of 1/5190 bases and 1/6315 bases after the first and second rounds of error correction, respectively.
  • TABLE 13
    Nucleic Acid Sequence
    Assembled 5′ATGACCATGATTACGGATTCACTGGCCG
    Gene, SEQ ID TCGTTTTACAACGTCGTGACTGGGAAAACC
    NO.: 67 CTGGCGTTACCCAACTTAATCGCCTTGCAG
    CACATCCCCCTTTCGCCAGCTGGCGTAATA
    GCGAAGAGGCCCGCACCGATCGCCCTTCCC
    AACAGTTGCGCAGCCTGAATGGCGAATGGC
    GCTTTGCCTGGTTTCCGGCACCAGAAGCGG
    TGCCGGAAAGCTGGCTGGAGTGCGATCTTC
    CTGAGGCCGATACTGTCGTCGTCCCCTCAA
    ACTGGCAGATGCACGGTTACGATGCGCCCA
    TCTACACCAACGTGACCTATCCCATTACGG
    TCAATCCGCCGTTTGTTCCCACGGAGAATC
    CGACGGGTTGTTACTCGCTCACATTTAATG
    TTGATGAAAGCTGGCTACAGGAAGGCCAGA
    CGCGAATTATTTTTGATGGCGTTAACTCGG
    CGTTTCATCTGTGGTGCAACGGGCGCTGGG
    TCGGTTACGGCCAGGACAGTCGTTTGCCGT
    CTGAATTTGACCTGAGCGCATTTTTACGCG
    CCGGAGAAAACCGCCTCGCGGTGATGGTGC
    TGCGCTGGAGTGACGGCAGTTATCTGGAAG
    ATCAGGATATGTGGCGGATGAGCGGCATTT
    TCCGTGACGTCTCGTTGCTGCATAAACCGA
    CTACACAAATCAGCGATTTCCATGTTGCCA
    CTCGCTTTAATGATGATTTCAGCCGCGCTG
    TACTGGAGGCTGAAGTTCAGATGTGCGGCG
    AGTTGCGTGACTACCTACGGGTAACAGTTT
    CTTTATGGCAGGGTGAAACGCAGGTCGCCA
    GCGGCACCGCGCCTTTCGGCGGTGAAATTA
    TCGATGAGCGTGGTGGTTATGCCGATCGCG
    TCACACTACGTCTGAACGTCGAAAACCCGA
    AACTGTGGAGCGCCGAAATCCCGAATCTCT
    ATC3′
    Assembly
    5′ATGACCATGATTACGGATTCACTGGCCG
    Oligonucleotide TCGTTTTACAACGTCGTGACTGGGAAAACC
    1, SEQ ID CTGGCGTTACCCAACTTAATCGCCTTGCAG
    NO.: 68 CACATCCCCCTTTCGCCAGCTGGCGTAATA
    GCGAAGAGGCCCGCACCGATCGCCCTTCCC
    AACAGTTGCGCAGCC
     3′
    Assembly 5′GATAGGTCACGTTGGTGTAGATGGGCGC
    Oligonucleotide ATCGTAACCGTGCATCTGCCAGTTTGAGGG
    2, SEQ ID GACGACGACAGTATCGGCCTCAGGAAGATC
    NO.: 69 GCACTCCAGCCAGCTTTCCGGCACCGCTTC
    TGGTGCCGGAAACCAGGCAAAGCGCCATTC
    GCCATTCAGGCTGCGCAACTGTTGGGA3′
    Assembly
    5′CCCATCTACACCAACGTGACCTATCCCA
    Oligonucleotide TTACGGTCAATCCGCCGTTTGTTCCCACGG
    3, SEQ ID AGAATCCGACGGGTTGTTACTCGCTCACAT
    NO.: 70 TTAATGTTGATGAAAGCTGGCTACAGGAAG
    GCCAGACGCGAATTATTTTTGATGGCGTTA
    ACTCGGCGTTTCATCTGTGGTGCAACGG3′
    Assembly
    5′GCCGCTCATCCGCCACATATCCTGATCT
    Oligonucleotide TCCAGATAACTGCCGTCACTCCAGCGCAGC
    4, SEQ ID ACCATCACCGCGAGGCGGTTTTCTCCGGCG
    NO.: 71 CGTAAAAATGCGCTCAGGTCAAATTCAGAC
    GGCAAACGACTGTCCTGGCCGTAACCGACC
    CAGCGCCCGTTGCACCACAGATGAAACG3′
    Assembly
    5′AGGATATGTGGCGGATGAGCGGCATTTT
    Oligonucleotide CCGTGACGTCTCGTTGCTGCATAAACCGAC
    5, SEQ ID TACACAAATCAGCGATTTCCATGTTGCCAC
    NO.: 72 TCGCTTTAATGATGATTTCAGCCGCGCTGT
    ACTGGAGGCTGAAGTTCAGATGTGCGGCGA
    GTTGCGTGACTACCTACGGGTAACAGTTT
    3′
    Assembly 5′GATAGAGATTCGGGATTTCGGCGCTCCA
    Oligonucleotide CAGTTTCGGGTTTTCGACGTTCAGACGTAG
    6, SEQ ID TGTGACGCGATCGGCATAACCACCACGCTC
    NO.: 73 ATCGATAATTTCACCGCCGAAAGGCGCGGT
    GCCGCTGGCGACCTGCGTTTCACCCTGCCA
    TAAAGAAACTGTTACCCGTAGGTAGTCACG
    3′
  • Example 7: Parallel Assembly of 240 Genes on Flat Plate
  • A substrate 1700 comprising 256 clusters each comprising 121 loci on a flat silicon plate was manufactured as shown in FIG. 17 . An expanded view of a cluster is shown in 1705 with 121 loci. Loci from 240 of the 256 clusters provided an attachment and support for the synthesis of oligonucleic acids having distinct sequences. Oligonucleic acid synthesis was performed by phosphoramidite chemistry using general methods from Table 5 in Example 4. Loci from 16 of the 256 clusters were control clusters. The distribution of the 29,040 unique oligonucleic acids synthesized (240×121) is shown in FIG. 18 . The distribution of unique oligonucleic acids synthesized in 4 representative clusters is shown in FIG. 19 . The error rate for each oligonucleic acid was determined using an Illumina MiSeq gene sequencer. The error rate distribution for the 29,040 unique oligonucleic acids is shown in FIG. 20 and averages around 1 in 500 bases, with some error rates as low as 1 in 800 bases. The error rate distribution for unique oligonucleic acids in four representative clusters is shown in FIG. 21 . The library of 29,040 unique oligonucleic acids was synthesized in less than 20 hours.
  • Oligonucleic acids synthesized within the loci of a cluster had overlapping sequences with one another so that when all oligonucleic acids synthesized within one cluster are pooled, they are ready for assembly into a larger nucleic acid or gene using PCA. Oligonucleic acids within a cluster were pooled and assembled using PCA reaction conditions similar to those described in Example 5. The 240 unique genes were synthesized in 3 business days, however, with 24 hour a day operation, the 240 unique genes could be synthesized in less time. The assembled genes from each of the 240 clusters were sequenced using an Illumina MiSeq gene sequencer. The read counts for the assembled genes are represented in FIG. 22 . The assembled gene products were visualized on a DNA gel as shown in FIGS. 23-26 . The genes synthesized ranged in size from 838 base pairs to 1470 base pairs. All 240 gene products were generated with their expected size. An output from the sequencing run is shown in FIG. 27 .
  • Example 8: Oligonucleic Acid Library Synthesis with Low Error Rate
  • A substrate comprising three-dimensional features with high surface area loci was manufactured according to the methods similar to that of Example 3. Each locus was manufactured to have a single comb shape. The substrate has a plurality of clusters corresponding to a plurality of wells, wherein each channel is 1.150 mm in diameter and includes 109 loci in the form of microchannels. FIG. 10C provides a depiction for a collection of microchannels/loci extending from a main channel. Microchannels of the substrate were functionalized and used as an attachment and support for the synthesis of distinct oligonucleic acids. A library of oligonucleic acids was synthesized on the substrate and subsequently gas cleaved from the surface for sequence analysis using an Illumina MiSeq.
  • Error mismatch rates for oligonucleic acids synthesized within each cluster were calculated (data not shown). Error rates were from 1 in 7,000 bases to as high as 1 in 100 bases. The average mismatch error rate was 1 in 586.82 bases.
  • Error deletion rates for oligonucleic acids synthesized within each cluster were calculated (data not shown). Error rates were from around 1 in 1,000 bases to around 1 in 2,000 bases. The average deletion error rate was 1 in 1,966.86 bases.
  • Insertion rates for oligonucleic acids synthesized within each cluster were calculated (data not shown). Error rates were from 1 in 2,700 bases to around 1 in 10,000 bases. The average insertion error rate was 1 in 4,740.03 bases.
  • The percentage of perfect oligonucleic acids synthesized (a perfect sequence being 100% identical to a preselected nucleic acid sequence) within each cluster were calculated (data not shown). The percentage of perfect sequences ranged from about 70% to about 93.54% perfect sequences. Overall more than 90% of the oligonucleic acids had perfect sequences.
  • Example 9: Linker Length Analysis
  • An oligonucleic acid library was synthesized as described in Example 8. Each oligonucleic acid synthesized on a locus of a cluster was tethered to the locus by a linker. Error rate as a function of base distance from substrate surface was analyzed and graphed as depicted in FIG. 28 . The lowest error rates correspond to oligonucleic acids with tether between about 12 and 25 bases from the surface.
  • Example 10: Parallel Synthesis of Distinct Oligonucleic Acids
  • Oligonucleic acids of various sequences and lengths were synthesized by phosphoramidite chemistry using methods as generally described in Table 5 of Example 4. Oligonucleic acids having lengths from 25 bases to 200 bases were synthesized within different clusters of a substrate. The synthesized oligonucleic acids were released from the surface, collected, and visualized by gel electrophoresis. FIG. 29 provides a captured image of the electrophoresis gel loaded with representative synthesized oligonucleic acids having lengths of 25, 50, 75, 100, 125, 150, 175 and 200 nucleotides.
  • As exemplified in FIG. 29 , the methods and substrates described herein allow for the simultaneous synthesis of a plurality of oligonucleic acids each having different sequences and, in some cases, different sequence lengths. In particular, oligonucleic acids having 200 bases were synthesized on, and removed from a substrate. These synthesized oligonucleic acids were released from the substrate and used in downstream processes, such as visualization by gel electrophoresis. Representative quantities of synthesized oligonucleic acids extracted from each cluster in this example ranged from 113 fmol to 344 fmol. Representative yields from each cluster ranged from 48 pmol/cm2 to 145 pmol/cm2.
  • While specific instances have been shown and described herein, it will be apparent to those skilled in the art that such instances are provided by way of example only. Numerous variations, changes, and substitutions will now occur to those skilled in the art without departing from the disclosed instances. It should be understood that various alternatives to the instances described herein may be employed in practicing the invention.

Claims (17)

What is claimed is:
1. A method for de novo oligonucleotide synthesis, comprising:
(a) providing predetermined sequences for a plurality of non-identical oligonucleotides;
(b) providing a device for synthesizing oligonucleotides, comprising:
(i) a solid substrate;
(ii) a main channel, wherein the main channel extends vertically into the solid substrate from an opening on a top side of the solid substrate; and
(iii) a plurality of microchannels, wherein each microchannel of the plurality of microchannels extends vertically from an opening on a bottom side of the solid substrate into the main channel, and wherein each microchannel of the plurality of microchannels comprises a fluid path having at least one turn of 45 degrees to 180 degrees in total, when viewed from a top view, and
wherein the device comprises more than 20,000 of the microchannels in total;
(c) adding a droplet of fluid comprising an extension reaction reagent specific to a microchannel;
(d) allowing sufficient time for an extension reaction step to occur; and
(e) repeating steps (c) and (d) until the plurality of non-identical oligonucleotides are synthesized.
2. The method of claim 1, wherein the fluid path comprises up to 10 turns in total.
3. The method of claim 1, wherein the at least one turn is 45, 90 or 180 degrees in total.
4. The method of claim 1, wherein the device comprises a total of at least 700,000 microchannels.
5. The method of claim 1, wherein the plurality of microchannels comprises 100 to 150 microchannels.
6. The method of claim 1, wherein the plurality of microchannels comprises up to 500 microchannels.
7. The method of claim 1, wherein a ratio of width to depth of a narrowest segment of each microchannel is from 0.5 to 0.01.
8. The method of claim 1, wherein each microchannel of the plurality of microchannels has a width of 30 um to 100 um.
9. The method of claim 1, wherein each microchannel of the plurality of microchannels has a depth of 10 um to 500 um.
10. The method of claim 1, wherein the plurality of microchannels has a higher surface energy than the main channel.
11. The method of claim 1, wherein the plurality of microchannels has a higher hydrophobicity than the main channel.
12. The method of claim 1, wherein the main channel has a width of 0.5 to 2 mm.
13. The method of claim 1, wherein the device comprises at least 50 main channels.
14. The method of claim 1, wherein the device comprises at least 500 main channels.
15. The method of claim 1, wherein the device comprises at least 5000 main channels.
16. The method of claim 1, wherein the solid substrate is in a form of a plate or a tape.
17. The method of claim 1, wherein the solid substrate comprises silicon, silicon dioxide, silicon nitride, nylon, nitrocellulose, polypropylene, or polydimethylsiloxane (PDMS).
US18/319,356 2015-04-21 2023-05-17 Devices and methods for oligonucleic acid library synthesis Abandoned US20230338913A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US18/319,356 US20230338913A1 (en) 2015-04-21 2023-05-17 Devices and methods for oligonucleic acid library synthesis

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US201562150795P 2015-04-21 2015-04-21
US201562220856P 2015-09-18 2015-09-18
US15/135,434 US9981239B2 (en) 2015-04-21 2016-04-21 Devices and methods for oligonucleic acid library synthesis
US15/960,319 US10744477B2 (en) 2015-04-21 2018-04-23 Devices and methods for oligonucleic acid library synthesis
US16/921,712 US11691118B2 (en) 2015-04-21 2020-07-06 Devices and methods for oligonucleic acid library synthesis
US18/319,356 US20230338913A1 (en) 2015-04-21 2023-05-17 Devices and methods for oligonucleic acid library synthesis

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US16/921,712 Continuation US11691118B2 (en) 2015-04-21 2020-07-06 Devices and methods for oligonucleic acid library synthesis

Publications (1)

Publication Number Publication Date
US20230338913A1 true US20230338913A1 (en) 2023-10-26

Family

ID=57143437

Family Applications (4)

Application Number Title Priority Date Filing Date
US15/135,434 Active 2036-05-03 US9981239B2 (en) 2015-04-21 2016-04-21 Devices and methods for oligonucleic acid library synthesis
US15/960,319 Active US10744477B2 (en) 2015-04-21 2018-04-23 Devices and methods for oligonucleic acid library synthesis
US16/921,712 Active 2037-06-27 US11691118B2 (en) 2015-04-21 2020-07-06 Devices and methods for oligonucleic acid library synthesis
US18/319,356 Abandoned US20230338913A1 (en) 2015-04-21 2023-05-17 Devices and methods for oligonucleic acid library synthesis

Family Applications Before (3)

Application Number Title Priority Date Filing Date
US15/135,434 Active 2036-05-03 US9981239B2 (en) 2015-04-21 2016-04-21 Devices and methods for oligonucleic acid library synthesis
US15/960,319 Active US10744477B2 (en) 2015-04-21 2018-04-23 Devices and methods for oligonucleic acid library synthesis
US16/921,712 Active 2037-06-27 US11691118B2 (en) 2015-04-21 2020-07-06 Devices and methods for oligonucleic acid library synthesis

Country Status (2)

Country Link
US (4) US9981239B2 (en)
WO (1) WO2016172377A1 (en)

Families Citing this family (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2943498T3 (en) 2013-08-05 2023-06-13 Twist Bioscience Corp de novo synthesized libraries
US10040048B1 (en) 2014-09-25 2018-08-07 Synthego Corporation Automated modular system and method for production of biopolymers
CA2975855A1 (en) 2015-02-04 2016-08-11 Twist Bioscience Corporation Compositions and methods for synthetic gene assembly
WO2016126882A1 (en) 2015-02-04 2016-08-11 Twist Bioscience Corporation Methods and devices for de novo oligonucleic acid assembly
US9981239B2 (en) 2015-04-21 2018-05-29 Twist Bioscience Corporation Devices and methods for oligonucleic acid library synthesis
GB2557529A (en) 2015-09-18 2018-06-20 Twist Bioscience Corp Oligonucleic acid variant libraries and synthesis thereof
WO2017053450A1 (en) 2015-09-22 2017-03-30 Twist Bioscience Corporation Flexible substrates for nucleic acid synthesis
WO2017095958A1 (en) 2015-12-01 2017-06-08 Twist Bioscience Corporation Functionalized surfaces and preparation thereof
CN109996876A (en) 2016-08-22 2019-07-09 特韦斯特生物科学公司 The nucleic acid library of de novo formation
WO2018057526A2 (en) 2016-09-21 2018-03-29 Twist Bioscience Corporation Nucleic acid based data storage
AU2017378492B2 (en) 2016-12-16 2022-06-16 Twist Bioscience Corporation Variant libraries of the immunological synapse and synthesis thereof
US11550939B2 (en) 2017-02-22 2023-01-10 Twist Bioscience Corporation Nucleic acid based data storage using enzymatic bioencryption
CA3056388A1 (en) 2017-03-15 2018-09-20 Twist Bioscience Corporation Variant libraries of the immunological synapse and synthesis thereof
WO2018217853A1 (en) * 2017-05-23 2018-11-29 Centrillion Technologies, Inc. Systems and methods to perform chemical synthesis on wafers
WO2018231872A1 (en) 2017-06-12 2018-12-20 Twist Bioscience Corporation Methods for seamless nucleic acid assembly
WO2018231864A1 (en) 2017-06-12 2018-12-20 Twist Bioscience Corporation Methods for seamless nucleic acid assembly
SG11202002194UA (en) 2017-09-11 2020-04-29 Twist Bioscience Corp Gpcr binding proteins and synthesis thereof
KR20240024357A (en) 2017-10-20 2024-02-23 트위스트 바이오사이언스 코포레이션 Heated nanowells for polynucleotide synthesis
KR20200106067A (en) 2018-01-04 2020-09-10 트위스트 바이오사이언스 코포레이션 DNA-based digital information storage
EP3814497A4 (en) 2018-05-18 2022-03-02 Twist Bioscience Corporation Polynucleotides, reagents, and methods for nucleic acid hybridization
US10704088B2 (en) * 2018-06-29 2020-07-07 Intel Corporation Massively parallel integrated circuit-based DNA synthesis devices, systems, and methods
EP3902816A4 (en) * 2018-12-26 2022-09-14 Twist Bioscience Corporation Highly accurate de novo polynucleotide synthesis
CN113785057A (en) 2019-02-26 2021-12-10 特韦斯特生物科学公司 Variant nucleic acid libraries for antibody optimization
CA3131689A1 (en) 2019-02-26 2020-09-03 Twist Bioscience Corporation Variant nucleic acid libraries for glp1 receptor
AU2020298294A1 (en) 2019-06-21 2022-02-17 Twist Bioscience Corporation Barcode-based nucleic acid sequence assembly
CN111018931A (en) * 2019-12-30 2020-04-17 江苏东玄基因科技有限公司 Automatic solid-phase synthesis system for oligomer
CN111748464B (en) * 2020-07-02 2022-04-12 浙江大学 Manufacturing method of digital PCR chip and digital PCR chip
EP4147712A1 (en) 2021-09-13 2023-03-15 OncoDNA Method to generate a double-stranded dna pool encoding neoantigens of a tumor of a patient
WO2023037000A2 (en) 2021-09-13 2023-03-16 Oncodna A rna vaccine comprising an rna pool generated from a double-stranded dna pool

Family Cites Families (935)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3549368A (en) 1968-07-02 1970-12-22 Ibm Process for improving photoresist adhesion
US3920714A (en) 1972-11-16 1975-11-18 Weber Heinrich Process for the production of polymeric hydrocarbons with reactive silyl side groups
GB1550867A (en) 1975-08-04 1979-08-22 Hughes Aircraft Co Positioning method and apparatus for fabricating microcircuit devices
US4415732A (en) 1981-03-27 1983-11-15 University Patents, Inc. Phosphoramidite compounds and processes
EP0090789A1 (en) 1982-03-26 1983-10-05 Monsanto Company Chemical DNA synthesis
US4994373A (en) 1983-01-27 1991-02-19 Enzo Biochem, Inc. Method and structures employing chemically-labelled polynucleotide probes
JPS59224123A (en) 1983-05-20 1984-12-17 Oki Electric Ind Co Ltd Alignment mark for wafer
US5118605A (en) 1984-10-16 1992-06-02 Chiron Corporation Polynucleotide determination with selectable cleavage sites
JPS61141761A (en) 1984-12-12 1986-06-28 Kanegafuchi Chem Ind Co Ltd Curable composition
US5242794A (en) 1984-12-13 1993-09-07 Applied Biosystems, Inc. Detection of specific sequences in nucleic acids
US6492107B1 (en) 1986-11-20 2002-12-10 Stuart Kauffman Process for obtaining DNA, RNA, peptides, polypeptides, or protein, by recombinant DNA technique
US4613398A (en) 1985-06-06 1986-09-23 International Business Machines Corporation Formation of etch-resistant resists through preferential permeation
US4981797A (en) 1985-08-08 1991-01-01 Life Technologies, Inc. Process of producing highly transformable cells and cells produced thereby
US4726877A (en) 1986-01-22 1988-02-23 E. I. Du Pont De Nemours And Company Methods of using photosensitive compositions containing microgels
US4808511A (en) 1987-05-19 1989-02-28 International Business Machines Corporation Vapor phase photoresist silylation process
JPH07113774B2 (en) 1987-05-29 1995-12-06 株式会社日立製作所 Pattern formation method
US4988617A (en) 1988-03-25 1991-01-29 California Institute Of Technology Method of detecting a nucleotide change in nucleic acids
US5700637A (en) 1988-05-03 1997-12-23 Isis Innovation Limited Apparatus and method for analyzing polynucleotide sequences and method of generating oligonucleotide arrays
ATE143696T1 (en) 1989-02-28 1996-10-15 Canon Kk PARTIALLY DOUBLE STRANDED OLIGONUCLEOTIDE AND METHOD FOR FORMING IT
US5556750A (en) 1989-05-12 1996-09-17 Duke University Methods and kits for fractionating a population of DNA molecules based on the presence or absence of a base-pair mismatch utilizing mismatch repair systems
US5459039A (en) 1989-05-12 1995-10-17 Duke University Methods for mapping genetic mutations
US6008031A (en) 1989-05-12 1999-12-28 Duke University Method of analysis and manipulation of DNA utilizing mismatch repair systems
US5102797A (en) 1989-05-26 1992-04-07 Dna Plant Technology Corporation Introduction of heterologous genes into bacteria using transposon flanked expression cassette and a binary vector system
US6919211B1 (en) 1989-06-07 2005-07-19 Affymetrix, Inc. Polypeptide arrays
US5242974A (en) 1991-11-22 1993-09-07 Affymax Technologies N.V. Polymer reversal on solid surfaces
US6040138A (en) 1995-09-15 2000-03-21 Affymetrix, Inc. Expression monitoring by hybridization to high density oligonucleotide arrays
US5527681A (en) 1989-06-07 1996-06-18 Affymax Technologies N.V. Immobilized molecular synthesis of systematically substituted compounds
US6309822B1 (en) 1989-06-07 2001-10-30 Affymetrix, Inc. Method for comparing copy number of nucleic acid sequences
US5143854A (en) 1989-06-07 1992-09-01 Affymax Technologies N.V. Large scale photolithographic solid phase synthesis of polypeptides and receptor binding screening thereof
US5744101A (en) 1989-06-07 1998-04-28 Affymax Technologies N.V. Photolabile nucleoside protecting groups
CA2036946C (en) 1990-04-06 2001-10-16 Kenneth V. Deugau Indexing linkers
US5494810A (en) 1990-05-03 1996-02-27 Cornell Research Foundation, Inc. Thermostable ligase-mediated DNA amplifications system for the detection of genetic disease
US6087482A (en) 1990-07-27 2000-07-11 Isis Pharmaceuticals, Inc. Heteroatomic oligonucleoside linkages
FI87886C (en) 1990-09-06 1993-03-10 Instrumentarium Oy Fasteners
AU8871891A (en) 1990-09-27 1992-04-28 Invitrogen Corporation Direct cloning of pcr amplified nucleic acids
GB9025236D0 (en) 1990-11-20 1991-01-02 Secr Defence Silicon-on porous-silicon;method of production
EP0562025B1 (en) 1990-12-06 2001-02-07 Affymetrix, Inc. (a Delaware Corporation) Compounds and their use in a binary synthesis strategy
DE69132843T2 (en) 1990-12-06 2002-09-12 Affymetrix Inc N D Ges D Staat Identification of nucleic acids in samples
US6582908B2 (en) 1990-12-06 2003-06-24 Affymetrix, Inc. Oligonucleotides
US5455166A (en) 1991-01-31 1995-10-03 Becton, Dickinson And Company Strand displacement amplification
US5137814A (en) 1991-06-14 1992-08-11 Life Technologies, Inc. Use of exo-sample nucleotides in gene cloning
US5449754A (en) 1991-08-07 1995-09-12 H & N Instruments, Inc. Generation of combinatorial libraries
US5474796A (en) 1991-09-04 1995-12-12 Protogene Laboratories, Inc. Method and apparatus for conducting an array of chemical reactions on a support surface
US5846717A (en) 1996-01-24 1998-12-08 Third Wave Technologies, Inc. Detection of nucleic acid sequences by invader-directed cleavage
US7150982B2 (en) 1991-09-09 2006-12-19 Third Wave Technologies, Inc. RNA detection assays
US6759226B1 (en) 2000-05-24 2004-07-06 Third Wave Technologies, Inc. Enzymes for the detection of specific nucleic acid sequences
US5994069A (en) 1996-01-24 1999-11-30 Third Wave Technologies, Inc. Detection of nucleic acids by multiple sequential invasive cleavages
US7045289B2 (en) 1991-09-09 2006-05-16 Third Wave Technologies, Inc. Detection of RNA Sequences
DE69233331T3 (en) 1991-11-22 2007-08-30 Affymetrix, Inc., Santa Clara Combinatorial Polymersynthesis Strategies
US5384261A (en) 1991-11-22 1995-01-24 Affymax Technologies N.V. Very large scale immobilized polymer synthesis using mechanically directed flow paths
US5573905A (en) 1992-03-30 1996-11-12 The Scripps Research Institute Encoded combinatorial chemical libraries
WO1993020236A1 (en) 1992-04-03 1993-10-14 Applied Biosystems, Inc. Probe composition and method
JP2553322Y2 (en) 1992-05-11 1997-11-05 サンデン株式会社 Filter feed mechanism of beverage brewing device
DE69310179T2 (en) 1992-07-31 1997-07-31 Behringwerke Ag METHOD FOR INTRODUCING DEFINED SEQUENCES AT THE 3 'END OF POLYNUCLEOTIDES
US5288514A (en) 1992-09-14 1994-02-22 The Regents Of The University Of California Solid phase and combinatorial synthesis of benzodiazepine compounds on a solid support
JP3176444B2 (en) 1992-10-01 2001-06-18 株式会社リコー Aqueous ink and recording method using the same
DE4241045C1 (en) 1992-12-05 1994-05-26 Bosch Gmbh Robert Process for anisotropic etching of silicon
US5368823A (en) 1993-02-11 1994-11-29 University Of Georgia Research Foundation, Inc. Automated synthesis of oligonucleotides
US5395753A (en) 1993-02-19 1995-03-07 Theratech, Inc. Method for diagnosing rheumatoid arthritis
DE69433010T2 (en) 1993-04-12 2004-06-09 Northwestern University, Evanston METHOD FOR PRESENTING OLIGONUCLEOTIDES
US7135312B2 (en) 1993-04-15 2006-11-14 University Of Rochester Circular DNA vectors for synthesis of RNA and DNA
US5455239A (en) 1993-08-05 1995-10-03 Merck & Co. Inc. 3-aryl of heteroaryl-7-heteroaralkylamido cephalosporin compounds, compositions and methods of use
US5482845A (en) 1993-09-24 1996-01-09 The Trustees Of Columbia University In The City Of New York Method for construction of normalized cDNA libraries
CN1039623C (en) 1993-10-22 1998-09-02 中国人民解放军军事医学科学院毒物药物研究所 Medicinal composite for preventing and curing kinetosis syndrome
US6893816B1 (en) 1993-10-28 2005-05-17 Houston Advanced Research Center Microfabricated, flowthrough porous apparatus for discrete detection of binding reactions
JP3488465B2 (en) 1993-10-28 2004-01-19 ヒューストン・アドバンスド・リサーチ・センター Microfabricated flow-through porosity device for separately detecting binding reactions
US6027877A (en) 1993-11-04 2000-02-22 Gene Check, Inc. Use of immobilized mismatch binding protein for detection of mutations and polymorphisms, purification of amplified DNA samples and allele identification
US5834252A (en) 1995-04-18 1998-11-10 Glaxo Group Limited End-complementary polymerase reaction
US6015880A (en) 1994-03-16 2000-01-18 California Institute Of Technology Method and substrate for performing multiple sequential reactions on a matrix
BR9507229A (en) 1994-03-29 1997-09-16 Novo Nordisk As Amylase detergent additive detergent composition use of a detergent and an amylase construction of a recombinant cell expression vector dna and process to produce amylase
US5514789A (en) 1994-04-21 1996-05-07 Barrskogen, Inc. Recovery of oligonucleotides by gas phase cleavage
SE512382C2 (en) 1994-04-26 2000-03-06 Ericsson Telefon Ab L M Device and method for placing elongate elements against or adjacent to a surface
CA2159830C (en) 1994-04-29 2001-07-03 Timothy M Woudenberg System for real time detection of nucleic acid amplification products
US6287850B1 (en) 1995-06-07 2001-09-11 Affymetrix, Inc. Bioarray chip reaction apparatus and its manufacture
AU689924B2 (en) 1994-06-23 1998-04-09 Affymax Technologies N.V. Photolabile compounds and methods for their use
US5641658A (en) 1994-08-03 1997-06-24 Mosaic Technologies, Inc. Method for performing amplification of nucleic acid with two primers bound to a single solid support
US5530516A (en) 1994-10-04 1996-06-25 Tamarack Scientific Co., Inc. Large-area projection exposure system
US6613560B1 (en) 1994-10-19 2003-09-02 Agilent Technologies, Inc. PCR microreactor for amplifying DNA using microquantities of sample fluid
US6635226B1 (en) 1994-10-19 2003-10-21 Agilent Technologies, Inc. Microanalytical device and use thereof for conducting chemical processes
US5556752A (en) 1994-10-24 1996-09-17 Affymetrix, Inc. Surface-bound, unimolecular, double-stranded DNA
AU4283196A (en) 1994-11-22 1996-06-17 Complex Fluid Systems, Inc. Non-aminic photoresist adhesion promoters for microelectronic applications
US5688642A (en) 1994-12-01 1997-11-18 The United States Of America As Represented By The Secretary Of The Navy Selective attachment of nucleic acid molecules to patterned self-assembled surfaces
US6017434A (en) 1995-05-09 2000-01-25 Curagen Corporation Apparatus and method for the generation, separation, detection, and recognition of biopolymer fragments
US5830655A (en) 1995-05-22 1998-11-03 Sri International Oligonucleotide sizing using cleavable primers
US5700642A (en) 1995-05-22 1997-12-23 Sri International Oligonucleotide sizing using immobilized cleavable primers
US5877280A (en) 1995-06-06 1999-03-02 The Mount Sinai School Of Medicine Of The City University Of New York Thermostable muts proteins
US6446682B1 (en) 1995-06-06 2002-09-10 James P. Viken Auto-loading fluid exchanger and method of use
US5707806A (en) 1995-06-07 1998-01-13 Genzyme Corporation Direct sequence identification of mutations by cleavage- and ligation-associated mutation-specific sequencing
US5780613A (en) 1995-08-01 1998-07-14 Northwestern University Covalent lock for self-assembled oligonucleotide constructs
US5712126A (en) 1995-08-01 1998-01-27 Yale University Analysis of gene expression by display of 3-end restriction fragments of CDNA
US5854033A (en) 1995-11-21 1998-12-29 Yale University Rolling circle replication reporter systems
US6537776B1 (en) 1999-06-14 2003-03-25 Diversa Corporation Synthetic ligation reassembly in directed evolution
US6352842B1 (en) 1995-12-07 2002-03-05 Diversa Corporation Exonucease-mediated gene assembly in directed evolution
JP2000501615A (en) 1995-12-15 2000-02-15 アマーシャム・ライフ・サイエンス・インコーポレーテッド Method using a mismatch repair system for detection and removal of mutant sequences generated during enzyme amplification
US5962271A (en) 1996-01-03 1999-10-05 Cloutech Laboratories, Inc. Methods and compositions for generating full-length cDNA having arbitrary nucleotide sequence at the 3'-end
US5976846A (en) 1996-01-13 1999-11-02 Passmore; Steven E. Method for multifragment in vivo cloning and mutation mapping
US6706471B1 (en) 1996-01-24 2004-03-16 Third Wave Technologies, Inc. Detection of nucleic acid sequences by invader-directed cleavage
US7122364B1 (en) 1998-03-24 2006-10-17 Third Wave Technologies, Inc. FEN endonucleases
US6090606A (en) 1996-01-24 2000-07-18 Third Wave Technologies, Inc. Cleavage agents
US5985557A (en) 1996-01-24 1999-11-16 Third Wave Technologies, Inc. Invasive cleavage of nucleic acids
US7432048B2 (en) 1996-11-29 2008-10-07 Third Wave Technologies, Inc. Reactions on a solid surface
US7527928B2 (en) 1996-11-29 2009-05-05 Third Wave Technologies, Inc. Reactions on a solid surface
US6274369B1 (en) 1996-02-02 2001-08-14 Invitrogen Corporation Method capable of increasing competency of bacterial cell transformation
US6013440A (en) 1996-03-11 2000-01-11 Affymetrix, Inc. Nucleic acid affinity columns
US6020481A (en) 1996-04-01 2000-02-01 The Perkin-Elmer Corporation Asymmetric benzoxanthene dyes
US6706875B1 (en) 1996-04-17 2004-03-16 Affyemtrix, Inc. Substrate preparation process
US5869245A (en) 1996-06-05 1999-02-09 Fox Chase Cancer Center Mismatch endonuclease and its use in identifying mutations in targeted polynucleotide strands
US5863801A (en) 1996-06-14 1999-01-26 Sarnoff Corporation Automated nucleic acid isolation
US6780982B2 (en) 1996-07-12 2004-08-24 Third Wave Technologies, Inc. Charge tags and the separation of nucleic acid molecules
US5853993A (en) 1996-10-21 1998-12-29 Hewlett-Packard Company Signal enhancement method and kit
WO1998022541A2 (en) 1996-11-08 1998-05-28 Ikonos Corporation Method for coating substrates
US5750672A (en) 1996-11-22 1998-05-12 Barrskogen, Inc. Anhydrous amine cleavage of oligonucleotides
CA2273204C (en) 1996-11-29 2008-10-14 Third Wave Technologies, Inc. Fen-1 endonucleases, mixtures and cleavage methods
WO1998029736A1 (en) 1996-12-31 1998-07-09 Genometrix Incorporated Multiplexed molecular analysis apparatus and method
EP1005529B1 (en) 1997-02-12 2005-04-27 Invitrogen Corporation Methods for lyophilizing competent cells
US5882496A (en) 1997-02-27 1999-03-16 The Regents Of The University Of California Porous silicon structures with high surface area/specific pore size
US6770748B2 (en) 1997-03-07 2004-08-03 Takeshi Imanishi Bicyclonucleoside and oligonucleotide analogue
US6028189A (en) 1997-03-20 2000-02-22 University Of Washington Solvent for oligonucleotide synthesis and methods of use
AU751956B2 (en) 1997-03-20 2002-09-05 University Of Washington Solvent for biopolymer synthesis, solvent microdroplets and methods of use
US6419883B1 (en) 1998-01-16 2002-07-16 University Of Washington Chemical synthesis using solvent microdroplets
DE69838724T2 (en) 1997-03-21 2008-10-30 Stratagene California, La Jolla POLYMERASE-IMPROVING FACTOR (PEF) -HOLDING EXTRACTS, PEF PROTEIN COMPLEXES, ISOLATED PEF PROTEIN, AND METHOD OF CLEANING AND IDENTIFICATION
US6969488B2 (en) 1998-05-22 2005-11-29 Solexa, Inc. System and apparatus for sequential processing of analytes
US5922593A (en) 1997-05-23 1999-07-13 Becton, Dickinson And Company Microbiological test panel and method therefor
WO1999000657A1 (en) 1997-06-26 1999-01-07 Perseptive Biosystems, Inc. High density sample holder for analysis of biological samples
GB9714716D0 (en) 1997-07-11 1997-09-17 Brax Genomics Ltd Characterising nucleic acids
US5989872A (en) 1997-08-12 1999-11-23 Clontech Laboratories, Inc. Methods and compositions for transferring DNA sequence information among vectors
US6027898A (en) 1997-08-18 2000-02-22 Transgenomic, Inc. Chromatographic method for mutation detection using mutation site specifically acting enzymes and chemicals
US6794499B2 (en) 1997-09-12 2004-09-21 Exiqon A/S Oligonucleotide analogues
US6136568A (en) 1997-09-15 2000-10-24 Hiatt; Andrew C. De novo polynucleotide synthesis using rolling templates
AU9393398A (en) 1997-09-16 1999-04-05 Egea Biosciences, Inc. Method for the complete chemical synthesis and assembly of genes and genomes
US6670127B2 (en) 1997-09-16 2003-12-30 Egea Biosciences, Inc. Method for assembly of a polynucleotide encoding a target polypeptide
US5976842A (en) 1997-10-30 1999-11-02 Clontech Laboratories, Inc. Methods and compositions for use in high fidelity polymerase chain reaction
US8182991B1 (en) 1997-11-26 2012-05-22 Third Wave Technologies, Inc. FEN-1 endonucleases, mixtures and cleavage methods
US6408308B1 (en) 1998-01-29 2002-06-18 Incyte Pharmaceuticals, Inc. System and method for generating, analyzing and storing normalized expression datasets from raw expression datasets derived from microarray includes nucleic acid probe sequences
US6287776B1 (en) 1998-02-02 2001-09-11 Signature Bioscience, Inc. Method for detecting and classifying nucleic acid hybridization
US6251588B1 (en) 1998-02-10 2001-06-26 Agilent Technologies, Inc. Method for evaluating oligonucleotide probe sequences
US6426184B1 (en) 1998-02-11 2002-07-30 The Regents Of The University Of Michigan Method and apparatus for chemical and biochemical reactions using photo-generated reagents
JP4698023B2 (en) 1998-02-23 2011-06-08 ウイスコンシン アラムニ リサーチ ファンデーション Method and apparatus for synthesizing DNA probe array
JP4493844B2 (en) 1998-03-25 2010-06-30 ランデグレン、ウルフ Rolling circle replication of padlock probe
US6284497B1 (en) 1998-04-09 2001-09-04 Trustees Of Boston University Nucleic acid arrays and methods of synthesis
US7321828B2 (en) 1998-04-13 2008-01-22 Isis Pharmaceuticals, Inc. System of components for preparing oligonucleotides
EP1071826B1 (en) 1998-04-13 2006-09-13 Isis Pharmaceuticals, Inc. Identification of genetic targets for modulation by oligonucleotides and generation of oligonucleotides for gene modulation
US6376285B1 (en) 1998-05-28 2002-04-23 Texas Instruments Incorporated Annealed porous silicon with epitaxial layer for SOI
US6274725B1 (en) 1998-06-02 2001-08-14 Isis Pharmaceuticals, Inc. Activators for oligonucleotide synthesis
US6130045A (en) 1998-06-11 2000-10-10 Clontech Laboratories, Inc. Thermostable polymerase
US6251595B1 (en) 1998-06-18 2001-06-26 Agilent Technologies, Inc. Methods and devices for carrying out chemical reactions
EP0967217B1 (en) 1998-06-22 2005-12-21 Affymetrix, Inc. (a California Corporation) Reagents and methods for solid phase synthesis and display
US7399844B2 (en) 1998-07-09 2008-07-15 Agilent Technologies, Inc. Method and reagents for analyzing the nucleotide sequence of nucleic acids
US6218118B1 (en) 1998-07-09 2001-04-17 Agilent Technologies, Inc. Method and mixture reagents for analyzing the nucleotide sequence of nucleic acids by mass spectrometry
US20030022207A1 (en) 1998-10-16 2003-01-30 Solexa, Ltd. Arrayed polynucleotides and their use in genome analysis
US6787308B2 (en) 1998-07-30 2004-09-07 Solexa Ltd. Arrayed biomolecules and their use in sequencing
US6222030B1 (en) 1998-08-03 2001-04-24 Agilent Technologies, Inc. Solid phase synthesis of oligonucleotides using carbonate protecting groups and alpha-effect nucleophile deprotection
US6951719B1 (en) 1999-08-11 2005-10-04 Proteus S.A. Process for obtaining recombined nucleotide sequences in vitro, libraries of sequences and sequences thus obtained
US6991922B2 (en) 1998-08-12 2006-01-31 Proteus S.A. Process for in vitro creation of recombinant polynucleotide sequences by oriented ligation
US6107038A (en) 1998-08-14 2000-08-22 Agilent Technologies Inc. Method of binding a plurality of chemicals on a substrate by electrophoretic self-assembly
US7097974B1 (en) 1998-08-28 2006-08-29 Febit Biotech Gmbh Support for a method for determining an analyte and a method for producing the support
US6258454B1 (en) 1998-09-01 2001-07-10 Agilent Technologies Inc. Functionalization of substrate surfaces with silane mixtures
US6461812B2 (en) 1998-09-09 2002-10-08 Agilent Technologies, Inc. Method and multiple reservoir apparatus for fabrication of biomolecular arrays
US6458583B1 (en) 1998-09-09 2002-10-01 Agilent Technologies, Inc. Method and apparatus for making nucleic acid arrays
US6287824B1 (en) 1998-09-15 2001-09-11 Yale University Molecular cloning using rolling circle amplification
AR021833A1 (en) 1998-09-30 2002-08-07 Applied Research Systems METHODS OF AMPLIFICATION AND SEQUENCING OF NUCLEIC ACID
US6399516B1 (en) 1998-10-30 2002-06-04 Massachusetts Institute Of Technology Plasma etch techniques for fabricating silicon structures from a substrate
US6309828B1 (en) 1998-11-18 2001-10-30 Agilent Technologies, Inc. Method and apparatus for fabricating replicate arrays of nucleic acid molecules
GB9900298D0 (en) 1999-01-07 1999-02-24 Medical Res Council Optical sorting method
US6376246B1 (en) 1999-02-05 2002-04-23 Maxygen, Inc. Oligonucleotide mediated nucleic acid recombination
AU2415200A (en) 1999-01-18 2000-08-01 Maxygen, Inc. Methods of populating data structures for use in evolutionary simulations
AU3210200A (en) 1999-01-19 2000-08-01 Maxygen, Inc. Oligonucleotide mediated nucleic acid recombination
US20070065838A1 (en) 1999-01-19 2007-03-22 Maxygen, Inc. Oligonucleotide mediated nucleic acid recombination
US6251685B1 (en) 1999-02-18 2001-06-26 Agilent Technologies, Inc. Readout method for molecular biological electronically addressable arrays
EP1728860B1 (en) 1999-02-19 2010-01-27 febit holding GmbH Method for producing polymers
ATE556149T1 (en) 1999-02-23 2012-05-15 Caliper Life Sciences Inc MANIPULATION OF MICROPARTICLES IN MICROFLUIDIC SYSTEMS
WO2000053617A1 (en) 1999-03-08 2000-09-14 Protogene Laboratories, Inc. Methods and compositions for economically synthesizing and assembling long dna sequences
US6824866B1 (en) 1999-04-08 2004-11-30 Affymetrix, Inc. Porous silica substrates for polymer synthesis and assays
US6284465B1 (en) 1999-04-15 2001-09-04 Agilent Technologies, Inc. Apparatus, systems and method for locating nucleic acids bound to surfaces
US6469156B1 (en) 1999-04-20 2002-10-22 The United States Of America As Represented By The Department Of Health And Human Services Rapid and sensitive method for detecting histoplasma capsulatum
US6221653B1 (en) 1999-04-27 2001-04-24 Agilent Technologies, Inc. Method of performing array-based hybridization assays using thermal inkjet deposition of sample fluids
US6518056B2 (en) 1999-04-27 2003-02-11 Agilent Technologies Inc. Apparatus, systems and method for assaying biological materials using an annular format
US6773676B2 (en) 1999-04-27 2004-08-10 Agilent Technologies, Inc. Devices for performing array hybridization assays and methods of using the same
US6300137B1 (en) 1999-04-28 2001-10-09 Agilent Technologies Inc. Method for synthesizing a specific, surface-bound polymer uniformly over an element of a molecular array
US6242266B1 (en) 1999-04-30 2001-06-05 Agilent Technologies Inc. Preparation of biopolymer arrays
US6323043B1 (en) 1999-04-30 2001-11-27 Agilent Technologies, Inc. Fabricating biopolymer arrays
US7276336B1 (en) 1999-07-22 2007-10-02 Agilent Technologies, Inc. Methods of fabricating an addressable array of biopolymer probes
NZ515189A (en) 1999-05-01 2004-05-28 Psimedica Ltd Derivatized porous silicon as a biomaterial to use as a material for componets in or on the surface of the human body
AU4703300A (en) 1999-05-06 2000-11-21 Mount Sinai School Of Medicine Of The City University Of New York, The Dna-based steganography
US7056661B2 (en) 1999-05-19 2006-06-06 Cornell Research Foundation, Inc. Method for sequencing nucleic acid molecules
AU5285800A (en) 1999-05-24 2000-12-12 Invitrogen Corporation Method for deblocking of labeled oligonucleotides
US6472147B1 (en) 1999-05-25 2002-10-29 The Scripps Research Institute Methods for display of heterodimeric proteins on filamentous phage using pVII and pIX, compositions, vectors and combinatorial libraries
US6132997A (en) 1999-05-28 2000-10-17 Agilent Technologies Method for linear mRNA amplification
US6815218B1 (en) 1999-06-09 2004-11-09 Massachusetts Institute Of Technology Methods for manufacturing bioelectronic devices
AU5882000A (en) 1999-06-22 2001-01-09 Invitrogen Corporation Improved primers and methods for the detection and discrimination of nucleic acids
DE19928410C2 (en) 1999-06-22 2002-11-28 Agilent Technologies Inc Device housing with a device for operating a laboratory microchip
US6709852B1 (en) 1999-06-22 2004-03-23 Invitrogen Corporation Rapid growing microorganisms for biotechnology applications
US6399394B1 (en) 1999-06-30 2002-06-04 Agilent Technologies, Inc. Testing multiple fluid samples with multiple biopolymer arrays
US6465183B2 (en) 1999-07-01 2002-10-15 Agilent Technologies, Inc. Multidentate arrays
US7504213B2 (en) 1999-07-09 2009-03-17 Agilent Technologies, Inc. Methods and apparatus for preparing arrays comprising features having degenerate biopolymers
US6461816B1 (en) 1999-07-09 2002-10-08 Agilent Technologies, Inc. Methods for controlling cross-hybridization in analysis of nucleic acid sequences
US6306599B1 (en) 1999-07-16 2001-10-23 Agilent Technologies Inc. Biopolymer arrays and their fabrication
US6346423B1 (en) 1999-07-16 2002-02-12 Agilent Technologies, Inc. Methods and compositions for producing biopolymeric arrays
US6201112B1 (en) 1999-07-22 2001-03-13 Agilent Technologies Inc. Method for 3′ end-labeling ribonucleic acids
US6180351B1 (en) 1999-07-22 2001-01-30 Agilent Technologies Inc. Chemical array fabrication with identifier
ATE542916T1 (en) 1999-08-18 2012-02-15 Illumina Inc METHODS FOR GENERATING OLIGONUCLEOTIDE SOLUTIONS
US6262490B1 (en) 1999-11-05 2001-07-17 Advanced Semiconductor Engineering, Inc. Substrate strip for use in packaging semiconductor chips
US6319674B1 (en) 1999-09-16 2001-11-20 Agilent Technologies, Inc. Methods for attaching substances to surfaces
US6743585B2 (en) 1999-09-16 2004-06-01 Agilent Technologies, Inc. Methods for preparing conjugates
US7211390B2 (en) 1999-09-16 2007-05-01 454 Life Sciences 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
US7078167B2 (en) 1999-09-17 2006-07-18 Agilent Technologies, Inc. Arrays having background features and methods for using the same
US7122303B2 (en) 1999-09-17 2006-10-17 Agilent Technologies, Inc. Arrays comprising background features that provide for a measure of a non-specific binding and methods for using the same
EP1218543A2 (en) 1999-09-29 2002-07-03 Solexa Ltd. Polynucleotide sequencing
DE19947495C2 (en) 1999-10-01 2003-05-28 Agilent Technologies Inc Microfluidic microchip
EP1235932A2 (en) 1999-10-08 2002-09-04 Protogene Laboratories, Inc. Method and apparatus for performing large numbers of reactions using array assembly
US6232072B1 (en) 1999-10-15 2001-05-15 Agilent Technologies, Inc. Biopolymer array inspection
US6451998B1 (en) 1999-10-18 2002-09-17 Agilent Technologies, Inc. Capping and de-capping during oligonucleotide synthesis
US6171797B1 (en) 1999-10-20 2001-01-09 Agilent Technologies Inc. Methods of making polymeric arrays
US6387636B1 (en) 1999-10-22 2002-05-14 Agilent Technologies, Inc. Method of shielding biosynthesis reactions from the ambient environment on an array
US7115423B1 (en) 1999-10-22 2006-10-03 Agilent Technologies, Inc. Fluidic structures within an array package
US6077674A (en) 1999-10-27 2000-06-20 Agilent Technologies Inc. Method of producing oligonucleotide arrays with features of high purity
US20010055761A1 (en) 1999-10-29 2001-12-27 Agilent Technologies Small scale dna synthesis using polymeric solid support with functionalized regions
US6406849B1 (en) 1999-10-29 2002-06-18 Agilent Technologies, Inc. Interrogating multi-featured arrays
US8268605B2 (en) 1999-10-29 2012-09-18 Agilent Technologies, Inc. Compositions and methods utilizing DNA polymerases
US6689319B1 (en) 1999-10-29 2004-02-10 Agilent Technologies, Ind. Apparatus for deposition and inspection of chemical and biological fluids
US6329210B1 (en) 1999-10-29 2001-12-11 Agilent Technologies, Inc. Method and apparatus for high volume polymer synthesis
US6428957B1 (en) 1999-11-08 2002-08-06 Agilent Technologies, Inc. Systems tools and methods of assaying biological materials using spatially-addressable arrays
US6440669B1 (en) 1999-11-10 2002-08-27 Agilent Technologies, Inc. Methods for applying small volumes of reagents
US7041445B2 (en) 1999-11-15 2006-05-09 Clontech Laboratories, Inc. Long oligonucleotide arrays
US6446642B1 (en) 1999-11-22 2002-09-10 Agilent Technologies, Inc. Method and apparatus to clean an inkjet reagent deposition device
US6582938B1 (en) 2001-05-11 2003-06-24 Affymetrix, Inc. Amplification of nucleic acids
US6800439B1 (en) 2000-01-06 2004-10-05 Affymetrix, Inc. Methods for improved array preparation
US20010039014A1 (en) 2000-01-11 2001-11-08 Maxygen, Inc. Integrated systems and methods for diversity generation and screening
EP1118661A1 (en) 2000-01-13 2001-07-25 Het Nederlands Kanker Instituut T cell receptor libraries
AU2001237965A1 (en) 2000-01-25 2001-08-07 Affymetrix, Inc. Method, system and computer software for providing a genomic web portal
US6587579B1 (en) 2000-01-26 2003-07-01 Agilent Technologies Inc. Feature quality in array fabrication
US6458526B1 (en) 2000-01-28 2002-10-01 Agilent Technologies, Inc. Method and apparatus to inhibit bubble formation in a fluid
US7198939B2 (en) 2000-01-28 2007-04-03 Agilent Technologies, Inc. Apparatus for interrogating an addressable array
US6406851B1 (en) 2000-01-28 2002-06-18 Agilent Technologies, Inc. Method for coating a substrate quickly and uniformly with a small volume of fluid
US6235483B1 (en) 2000-01-31 2001-05-22 Agilent Technologies, Inc. Methods and kits for indirect labeling of nucleic acids
GB0002389D0 (en) 2000-02-02 2000-03-22 Solexa Ltd Molecular arrays
US6403314B1 (en) 2000-02-04 2002-06-11 Agilent Technologies, Inc. Computational method and system for predicting fragmented hybridization and for identifying potential cross-hybridization
US6833450B1 (en) 2000-03-17 2004-12-21 Affymetrix, Inc. Phosphite ester oxidation in nucleic acid array preparation
US6365355B1 (en) 2000-03-28 2002-04-02 The Regents Of The University Of California Chimeric proteins for detection and quantitation of DNA mutations, DNA sequence variations, DNA damage and DNA mismatches
US20020025561A1 (en) 2000-04-17 2002-02-28 Hodgson Clague Pitman Vectors for gene-self-assembly
US7776021B2 (en) 2000-04-28 2010-08-17 The Charles Stark Draper Laboratory Micromachined bilayer unit for filtration of small molecules
US6716634B1 (en) 2000-05-31 2004-04-06 Agilent Technologies, Inc. Increasing ionization efficiency in mass spectrometry
US7163660B2 (en) 2000-05-31 2007-01-16 Infineon Technologies Ag Arrangement for taking up liquid analytes
US6664112B2 (en) 2000-06-02 2003-12-16 Blue Heron Biotechnology, Inc. Methods for improving the sequence fidelity of synthetic double-stranded oligonucleotides
US6686193B2 (en) 2000-07-10 2004-02-03 Vertex Pharmaceuticals, Inc. High throughput method and system for screening candidate compounds for activity against target ion channels
EP1322780A4 (en) 2000-07-27 2005-08-03 Univ Australian Combinatorial probes and uses therefor
US7135565B2 (en) 2000-07-28 2006-11-14 Agilent Technologies, Inc. Synthesis of polynucleotides using combined oxidation/deprotection chemistry
US6599693B1 (en) 2000-07-31 2003-07-29 Agilent Technologies Inc. Array fabrication
US6613893B1 (en) 2000-07-31 2003-09-02 Agilent Technologies Inc. Array fabrication
US6890760B1 (en) 2000-07-31 2005-05-10 Agilent Technologies, Inc. Array fabrication
DE60114525T2 (en) 2000-07-31 2006-07-20 Agilent Technologies Inc., A Delaware Corp., Palo Alto Array-based methods for the synthesis of nucleic acid mixtures
US7205400B2 (en) 2000-07-31 2007-04-17 Agilent Technologies, Inc. Array fabrication
GB0018876D0 (en) 2000-08-01 2000-09-20 Applied Research Systems Method of producing polypeptides
US20020132308A1 (en) 2000-08-24 2002-09-19 Mpep @ Page 300-M Novel constructs and their use in metabolic pathway engineering
CA2421266A1 (en) 2000-09-08 2002-03-14 Richard T. Pon Linker phosphoramidites for oligonucleotide synthesis
US6966945B1 (en) 2000-09-20 2005-11-22 Goodrich Corporation Inorganic matrix compositions, composites and process of making the same
WO2002027029A2 (en) 2000-09-27 2002-04-04 Lynx Therapeutics, Inc. Method for determining relative abundance of nucleic acid sequences
NO20004869D0 (en) 2000-09-28 2000-09-28 Torbjoern Rognes Method for fast optimal local sequence alignment using parallel processing
US7097809B2 (en) 2000-10-03 2006-08-29 California Institute Of Technology Combinatorial synthesis system
EP1330306A2 (en) 2000-10-10 2003-07-30 BioTrove, Inc. Apparatus for assay, synthesis and storage, and methods of manufacture, use, and manipulation thereof
US6693187B1 (en) 2000-10-17 2004-02-17 Lievre Cornu Llc Phosphinoamidite carboxlates and analogs thereof in the synthesis of oligonucleotides having reduced internucleotide charge
DE10051396A1 (en) * 2000-10-17 2002-04-18 Febit Ferrarius Biotech Gmbh An integrated synthesis and identification of an analyte, comprises particles immobilized at a carrier to be coupled to receptors in a structured pattern to give receptor arrays for biochemical reactions
WO2002033669A1 (en) 2000-10-18 2002-04-25 Ultra Proizvodnja Elektronskih Naprav D.O.O. System for payment data exchange and payment terminal device used therein
EP1203945B1 (en) 2000-10-26 2006-12-20 Agilent Technologies, Inc. (a Delaware corporation) Microarray
US6905816B2 (en) 2000-11-27 2005-06-14 Intelligent Medical Devices, Inc. Clinically intelligent diagnostic devices and methods
US20020155439A1 (en) 2000-12-04 2002-10-24 Ana Rodriguez Method for generating a library of mutant oligonucleotides using the linear cyclic amplification reaction
US20040253242A1 (en) 2000-12-05 2004-12-16 Bowdish Katherine S. Rationally designed antibodies
US6768005B2 (en) 2000-12-20 2004-07-27 Avecia Limited Process
JP2004517089A (en) 2000-12-05 2004-06-10 アベシア・リミテッド Method for preparing phosphorothioate oligonucleotides
DE10060433B4 (en) 2000-12-05 2006-05-11 Hahn-Schickard-Gesellschaft für angewandte Forschung e.V. Method for producing a fluid component, fluid component and analysis device
US6660475B2 (en) 2000-12-15 2003-12-09 New England Biolabs, Inc. Use of site-specific nicking endonucleases to create single-stranded regions and applications thereof
AUPR259301A0 (en) 2001-01-18 2001-02-15 Polymerat Pty Ltd Polymers having co-continuous architecture
DE60227361D1 (en) 2001-01-19 2008-08-14 Centocor Inc COMPUTER MEDIATED ASSEMBLY OF POLYNUCLEOTIDES ENCODING A TARGETED POLYPEPTIDE
US6958217B2 (en) 2001-01-24 2005-10-25 Genomic Expression Aps Single-stranded polynucleotide tags
US7027930B2 (en) 2001-01-31 2006-04-11 Agilent Technologies, Inc. Reading chemical arrays
US6879915B2 (en) 2001-01-31 2005-04-12 Agilent Technologies, Inc. Chemical array fabrication and use
US7166258B2 (en) 2001-01-31 2007-01-23 Agilent Technologies, Inc. Automation-optimized microarray package
US20020164824A1 (en) 2001-02-16 2002-11-07 Jianming Xiao Method and apparatus based on bundled capillaries for high throughput screening
US6660338B1 (en) 2001-03-08 2003-12-09 Agilent Technologies, Inc. Functionalization of substrate surfaces with silane mixtures
WO2002072864A2 (en) 2001-03-08 2002-09-19 Applera Corporation Reagents for oligonucleotide cleavage and deprotection
US7211654B2 (en) 2001-03-14 2007-05-01 Regents Of The University Of Michigan Linkers and co-coupling agents for optimization of oligonucleotide synthesis and purification on solid supports
EP1370690B1 (en) 2001-03-16 2012-03-14 Kalim Mir Arrays and methods of use
US6610978B2 (en) 2001-03-27 2003-08-26 Agilent Technologies, Inc. Integrated sample preparation, separation and introduction microdevice for inductively coupled plasma mass spectrometry
US7208322B2 (en) 2001-04-02 2007-04-24 Agilent Technologies, Inc. Sensor surfaces for detecting analytes
US20030022240A1 (en) 2001-04-17 2003-01-30 Peizhi Luo Generation and affinity maturation of antibody library in silico
US6943036B2 (en) 2001-04-30 2005-09-13 Agilent Technologies, Inc. Error detection in chemical array fabrication
AU2002340641A1 (en) 2001-05-03 2002-11-18 Sigma-Genosys, Ltd. Methods for assembling protein microarrays
CA2447240C (en) 2001-05-18 2013-02-19 Wisconsin Alumni Research Foundation Method for the synthesis of dna sequences
WO2002094846A2 (en) 2001-05-22 2002-11-28 Parallel Synthesis Technologies, Inc. Method for in situ, on-chip chemical synthesis
US6880576B2 (en) 2001-06-07 2005-04-19 Nanostream, Inc. Microfluidic devices for methods development
US6649348B2 (en) 2001-06-29 2003-11-18 Agilent Technologies Inc. Methods for manufacturing arrays
US6613523B2 (en) 2001-06-29 2003-09-02 Agilent Technologies, Inc. Method of DNA sequencing using cleavable tags
US20040161741A1 (en) 2001-06-30 2004-08-19 Elazar Rabani Novel compositions and processes for analyte detection, quantification and amplification
US6989267B2 (en) 2001-07-02 2006-01-24 Agilent Technologies, Inc. Methods of making microarrays with substrate surfaces having covalently bound polyelectrolyte films
US6753145B2 (en) 2001-07-05 2004-06-22 Agilent Technologies, Inc. Buffer composition and method for hybridization of microarrays on adsorbed polymer siliceous surfaces
US7205399B1 (en) 2001-07-06 2007-04-17 Sirna Therapeutics, Inc. Methods and reagents for oligonucleotide synthesis
US7314599B2 (en) 2001-07-17 2008-01-01 Agilent Technologies, Inc. Paek embossing and adhesion for microfluidic devices
US6702256B2 (en) 2001-07-17 2004-03-09 Agilent Technologies, Inc. Flow-switching microdevice
US7128876B2 (en) 2001-07-17 2006-10-31 Agilent Technologies, Inc. Microdevice and method for component separation in a fluid
US20030108903A1 (en) 2001-07-19 2003-06-12 Liman Wang Multiple word DNA computing on surfaces
DE60232523D1 (en) 2001-07-26 2009-07-16 Stratagene California MULTI-JOBS Mutagenesis
US20030130827A1 (en) 2001-08-10 2003-07-10 Joerg Bentzien Protein design automation for protein libraries
US7371580B2 (en) 2001-08-24 2008-05-13 Agilent Technologies, Inc. Use of unstructured nucleic acids in assaying nucleic acid molecules
US6682702B2 (en) 2001-08-24 2004-01-27 Agilent Technologies, Inc. Apparatus and method for simultaneously conducting multiple chemical reactions
JP2003101204A (en) 2001-09-25 2003-04-04 Nec Kansai Ltd Wiring substrate, method of manufacturing the same, and electronic component
US20030082618A1 (en) 2001-10-15 2003-05-01 Guangshan Li Methods for detecting genetic aberrations
US6902921B2 (en) 2001-10-30 2005-06-07 454 Corporation Sulfurylase-luciferase fusion proteins and thermostable sulfurylase
US20050124022A1 (en) 2001-10-30 2005-06-09 Maithreyan Srinivasan Novel sulfurylase-luciferase fusion proteins and thermostable sulfurylase
US6858720B2 (en) 2001-10-31 2005-02-22 Agilent Technologies, Inc. Method of synthesizing polynucleotides using ionic liquids
US7524950B2 (en) 2001-10-31 2009-04-28 Agilent Technologies, Inc. Uses of cationic salts for polynucleotide synthesis
US6852850B2 (en) 2001-10-31 2005-02-08 Agilent Technologies, Inc. Use of ionic liquids for fabrication of polynucleotide arrays
US20030087298A1 (en) 2001-11-02 2003-05-08 Roland Green Detection of hybridization on oligonucleotide microarray through covalently labeling microarray probe
ATE509272T1 (en) 2001-11-09 2011-05-15 3Dbiosurfaces Technologies Llc SUBSTRATES WITH HIGH SURFACE AREA FOR MICROARRAYS AND METHOD FOR PRODUCING SAME
US7482118B2 (en) 2001-11-15 2009-01-27 Third Wave Technologies, Inc. Endonuclease-substrate complexes
ATE414767T1 (en) 2001-11-22 2008-12-15 Sloning Biotechnology Gmbh NUCLEIC ACID LINKERS AND THEIR USE IN GENE SYNTHESIS
US20030099952A1 (en) 2001-11-26 2003-05-29 Roland Green Microarrays with visible pattern detection
US20030143605A1 (en) 2001-12-03 2003-07-31 Si Lok Methods for the selection and cloning of nucleic acid molecules free of unwanted nucleotide sequence alterations
US6927029B2 (en) 2001-12-03 2005-08-09 Agilent Technologies, Inc. Surface with tethered polymeric species for binding biomolecules
US6838888B2 (en) 2001-12-13 2005-01-04 Agilent Technologies, Inc. Flow cell humidity sensor system
WO2003054232A2 (en) 2001-12-13 2003-07-03 Blue Heron Biotechnology, Inc. Methods for removal of double-stranded oligonucleotides containing sequence errors using mismatch recognition proteins
US7932070B2 (en) 2001-12-21 2011-04-26 Agilent Technologies, Inc. High fidelity DNA polymerase compositions and uses therefor
US7282183B2 (en) 2001-12-24 2007-10-16 Agilent Technologies, Inc. Atmospheric control in reaction chambers
US6790620B2 (en) 2001-12-24 2004-09-14 Agilent Technologies, Inc. Small volume chambers
US6846454B2 (en) 2001-12-24 2005-01-25 Agilent Technologies, Inc. Fluid exit in reaction chambers
US7025324B1 (en) 2002-01-04 2006-04-11 Massachusetts Institute Of Technology Gating apparatus and method of manufacture
WO2003057924A1 (en) 2002-01-04 2003-07-17 Board Of Regents, The University Of Texas System Proofreading, error deletion, and ligation method for synthesis of high-fidelity polynucleotide sequences
WO2003060084A2 (en) 2002-01-14 2003-07-24 Diversa Corporation Methods for making polynucleotides and purifying double-stranded polynucleotides
US20040009498A1 (en) 2002-01-14 2004-01-15 Diversa Corporation Chimeric antigen binding molecules and methods for making and using them
US6673552B2 (en) 2002-01-14 2004-01-06 Diversa Corporation Methods for purifying annealed double-stranded oligonucleotides lacking base pair mismatches or nucleotide gaps
US7141368B2 (en) 2002-01-30 2006-11-28 Agilent Technologies, Inc. Multi-directional deposition in array fabrication
US7422851B2 (en) 2002-01-31 2008-09-09 Nimblegen Systems, Inc. Correction for illumination non-uniformity during the synthesis of arrays of oligomers
US7157229B2 (en) 2002-01-31 2007-01-02 Nimblegen Systems, Inc. Prepatterned substrate for optical synthesis of DNA probes
US20040126757A1 (en) 2002-01-31 2004-07-01 Francesco Cerrina Method and apparatus for synthesis of arrays of DNA probes
US7037659B2 (en) 2002-01-31 2006-05-02 Nimblegen Systems Inc. Apparatus for constructing DNA probes having a prismatic and kaleidoscopic light homogenizer
US7083975B2 (en) 2002-02-01 2006-08-01 Roland Green Microarray synthesis instrument and method
US20030148291A1 (en) 2002-02-05 2003-08-07 Karla Robotti Method of immobilizing biologically active molecules for assay purposes in a microfluidic format
US6728129B2 (en) 2002-02-19 2004-04-27 The Regents Of The University Of California Multistate triple-decker dyads in three distinct architectures for information storage applications
US6958119B2 (en) 2002-02-26 2005-10-25 Agilent Technologies, Inc. Mobile phase gradient generation microfluidic device
US6929951B2 (en) 2002-02-28 2005-08-16 Agilent Technologies, Inc. Method and system for molecular array scanner calibration
US6770892B2 (en) 2002-02-28 2004-08-03 Agilent Technologies, Inc. Method and system for automated focus-distance determination for molecular array scanners
US6914229B2 (en) 2002-02-28 2005-07-05 Agilent Technologies, Inc. Signal offset for prevention of data clipping in a molecular array scanner
US20050084907A1 (en) 2002-03-01 2005-04-21 Maxygen, Inc. Methods, systems, and software for identifying functional biomolecules
US6919181B2 (en) 2002-03-25 2005-07-19 Agilent Technologies, Inc. Methods for generating ligand arrays
US20030228602A1 (en) 2002-04-01 2003-12-11 Blue Heron Biotechnology, Inc. Solid phase methods for polynucleotide production
EP1350853A1 (en) 2002-04-05 2003-10-08 ID-Lelystad, Instituut voor Dierhouderij en Diergezondheid B.V. Detection of polymorphisms
US6773888B2 (en) 2002-04-08 2004-08-10 Affymetrix, Inc. Photoactivatable silane compounds and methods for their synthesis and use
DE60322210D1 (en) 2002-04-22 2008-08-28 Genencor Int METHOD FOR GENERATING A LIBRARY FROM BACTERIA CLONES WITH DIFFERENT GENE EXPRESSION LEVELS
GB0209539D0 (en) 2002-04-26 2002-06-05 Avecia Ltd Monomer Polymer and process
US7125523B2 (en) 2002-04-29 2006-10-24 Agilent Technologies, Inc. Holders for arrays
US6946285B2 (en) 2002-04-29 2005-09-20 Agilent Technologies, Inc. Arrays with elongated features
US7094537B2 (en) 2002-04-30 2006-08-22 Agilent Technologies, Inc. Micro arrays with structured and unstructured probes
US6621076B1 (en) 2002-04-30 2003-09-16 Agilent Technologies, Inc. Flexible assembly for transporting sample fluids into a mass spectrometer
WO2003093504A1 (en) 2002-05-06 2003-11-13 Noxxon Pharma Ag Method for amplifying nucleic acids
US20030211478A1 (en) 2002-05-08 2003-11-13 Gentel Corporation Transcription factor profiling on a solid surface
US7221785B2 (en) 2002-05-21 2007-05-22 Agilent Technologies, Inc. Method and system for measuring a molecular array background signal from a continuous background region of specified size
US7273730B2 (en) 2002-05-24 2007-09-25 Invitrogen Corporation Nested PCR employing degradable primers
US7888106B2 (en) 2002-05-24 2011-02-15 Roche Nimblegen, Inc. Microarrays and method for running hybridization reaction for multiple samples on a single microarray
US7537936B2 (en) 2002-05-31 2009-05-26 Agilent Technologies, Inc. Method of testing multiple fluid samples with multiple biopolymer arrays
US6789965B2 (en) 2002-05-31 2004-09-14 Agilent Technologies, Inc. Dot printer with off-axis loading
US7078505B2 (en) 2002-06-06 2006-07-18 Agilent Technologies, Inc. Manufacture of arrays with varying deposition parameters
US7371348B2 (en) 2002-06-14 2008-05-13 Agilent Technologies Multiple array format
US7351379B2 (en) 2002-06-14 2008-04-01 Agilent Technologies, Inc. Fluid containment structure
US7919308B2 (en) 2002-06-14 2011-04-05 Agilent Technologies, Inc. Form in place gaskets for assays
US6939673B2 (en) 2002-06-14 2005-09-06 Agilent Technologies, Inc. Manufacture of arrays with reduced error impact
US7220573B2 (en) 2002-06-21 2007-05-22 Agilent Technologies, Inc. Array assay devices and methods of using the same
US6713262B2 (en) 2002-06-25 2004-03-30 Agilent Technologies, Inc. Methods and compositions for high throughput identification of protein/nucleic acid binding pairs
US7894998B2 (en) 2002-06-26 2011-02-22 Agilent Technologies, Inc. Method for identifying suitable nucleic acid probe sequences for use in nucleic acid arrays
US7202358B2 (en) 2002-07-25 2007-04-10 Agilent Technologies, Inc. Methods for producing ligand arrays
US7452712B2 (en) 2002-07-30 2008-11-18 Applied Biosystems Inc. Sample block apparatus and method of maintaining a microcard on a sample block
US7101508B2 (en) 2002-07-31 2006-09-05 Agilent Technologies, Inc. Chemical array fabrication errors
US6835938B2 (en) 2002-07-31 2004-12-28 Agilent Technologies, Inc. Biopolymer array substrate thickness dependent automated focus-distance determination method for biopolymer array scanners
US7153689B2 (en) 2002-08-01 2006-12-26 Agilent Technologies, Inc. Apparatus and methods for cleaning and priming droplet dispensing devices
US8946387B2 (en) 2002-08-14 2015-02-03 Macrogenics, Inc. FcγRIIB specific antibodies and methods of use thereof
US7205128B2 (en) 2002-08-16 2007-04-17 Agilent Technologies, Inc. Method for synthesis of the second strand of cDNA
US7563600B2 (en) 2002-09-12 2009-07-21 Combimatrix Corporation Microarray synthesis and assembly of gene-length polynucleotides
JP2006517090A (en) 2002-09-26 2006-07-20 コーサン バイオサイエンシーズ, インコーポレイテッド Synthetic gene
WO2004029586A1 (en) 2002-09-27 2004-04-08 Nimblegen Systems, Inc. Microarray with hydrophobic barriers
US7482170B2 (en) 2002-09-30 2009-01-27 Roche Nimblegen, Inc. Parallel loading of arrays
DE10393431T5 (en) 2002-10-01 2005-11-17 Nimblegen Systems, Inc., Madison Microarrays with multiple oligonucleotides in single array features
US7129075B2 (en) 2002-10-18 2006-10-31 Transgenomic, Inc. Isolated CEL II endonuclease
US8283148B2 (en) 2002-10-25 2012-10-09 Agilent Technologies, Inc. DNA polymerase compositions for quantitative PCR and methods thereof
WO2004039953A2 (en) 2002-10-28 2004-05-13 Xeotron Corporation Array oligomer synthesis and use.
US7422911B2 (en) 2002-10-31 2008-09-09 Agilent Technologies, Inc. Composite flexible array substrate having flexible support
US7390457B2 (en) 2002-10-31 2008-06-24 Agilent Technologies, Inc. Integrated microfluidic array device
US7364896B2 (en) 2002-10-31 2008-04-29 Agilent Technologies, Inc. Test strips including flexible array substrates and method of hybridization
US7402279B2 (en) 2002-10-31 2008-07-22 Agilent Technologies, Inc. Device with integrated microfluidic and electronic components
US7629120B2 (en) 2002-10-31 2009-12-08 Rice University Method for assembling PCR fragments of DNA
US6976384B2 (en) 2002-10-31 2005-12-20 Nanostream, Inc. Parallel detection chromatography systems
US20040086892A1 (en) 2002-11-06 2004-05-06 Crothers Donald M. Universal tag assay
US7029854B2 (en) 2002-11-22 2006-04-18 Agilent Technologies, Inc. Methods designing multiple mRNA transcript nucleic acid probe sequences for use in nucleic acid arrays
US7062385B2 (en) 2002-11-25 2006-06-13 Tufts University Intelligent electro-optical nucleic acid-based sensor array and method for detecting volatile compounds in ambient air
AU2003298816C1 (en) 2002-12-02 2010-12-16 Amgen Fremont, Inc. Antibodies directed to Tumor Necrosis Factor and uses thereof
US20040110133A1 (en) 2002-12-06 2004-06-10 Affymetrix, Inc. Functionated photoacid generator for biological microarray synthesis
US7879580B2 (en) 2002-12-10 2011-02-01 Massachusetts Institute Of Technology Methods for high fidelity production of long nucleic acid molecules
US7932025B2 (en) 2002-12-10 2011-04-26 Massachusetts Institute Of Technology Methods for high fidelity production of long nucleic acid molecules with error control
US20060076482A1 (en) 2002-12-13 2006-04-13 Hobbs Steven E High throughput systems and methods for parallel sample analysis
US6987263B2 (en) 2002-12-13 2006-01-17 Nanostream, Inc. High throughput systems and methods for parallel sample analysis
US7247337B1 (en) 2002-12-16 2007-07-24 Agilent Technologies, Inc. Method and apparatus for microarray fabrication
US20040191810A1 (en) 2002-12-17 2004-09-30 Affymetrix, Inc. Immersed microarrays in conical wells
GB0229443D0 (en) 2002-12-18 2003-01-22 Avecia Ltd Process
US7960157B2 (en) 2002-12-20 2011-06-14 Agilent Technologies, Inc. DNA polymerase blends and uses thereof
DE10260805A1 (en) 2002-12-23 2004-07-22 Geneart Gmbh Method and device for optimizing a nucleotide sequence for expression of a protein
EP1576192B1 (en) 2002-12-23 2014-10-29 Agilent Technologies, Inc. Comparative genomic hybridization assays using immobilized oligonucleotide features and compositions for practicing the same
AU2003298235A1 (en) 2002-12-23 2004-07-22 Febit Ag Intramolecular triplet-sensitized o-nitrophenylethyl photoprotective groups
JP4511943B2 (en) 2002-12-23 2010-07-28 ワイス エルエルシー Antibody against PD-1 and use thereof
US7372982B2 (en) 2003-01-14 2008-05-13 Agilent Technologies, Inc. User interface for molecular array feature analysis
US6809277B2 (en) 2003-01-22 2004-10-26 Agilent Technologies, Inc. Method for registering a deposited material with channel plate channels, and switch produced using same
ES2338654T5 (en) 2003-01-29 2017-12-11 454 Life Sciences Corporation Pearl emulsion nucleic acid amplification
US8073626B2 (en) 2003-01-31 2011-12-06 Agilent Technologies, Inc. Biopolymer array reading
US7202264B2 (en) 2003-01-31 2007-04-10 Isis Pharmaceuticals, Inc. Supports for oligomer synthesis
US6950756B2 (en) 2003-02-05 2005-09-27 Agilent Technologies, Inc. Rearrangement of microarray scan images to form virtual arrays
US7413709B2 (en) 2003-02-12 2008-08-19 Agilent Technologies, Inc. PAEK-based microfluidic device with integrated electrospray emitter
GB2398383B (en) 2003-02-12 2005-03-09 Global Genomics Ab Method and means for nucleic acid sequencing
US7244513B2 (en) 2003-02-21 2007-07-17 Nano-Proprietary, Inc. Stain-etched silicon powder
US7070932B2 (en) 2003-02-25 2006-07-04 Agilent Technologies, Inc. Methods and devices for detecting printhead misalignment of an in situ polymeric array synthesis device
US7252938B2 (en) 2003-02-25 2007-08-07 Agilent Technologies, Inc. Methods and devices for producing a polymer at a location of a substrate
US6977223B2 (en) 2003-03-07 2005-12-20 Massachusetts Institute Of Technology Three dimensional microfabrication
JP2004268394A (en) 2003-03-07 2004-09-30 Canon Inc Ink jet recorder and its controlling method
US20050053968A1 (en) 2003-03-31 2005-03-10 Council Of Scientific And Industrial Research Method for storing information in DNA
WO2004090170A1 (en) 2003-04-02 2004-10-21 Blue Heron Biotechnology, Inc. Error reduction in automated gene synthesis
US7534561B2 (en) 2003-04-02 2009-05-19 Agilent Technologies, Inc. Nucleic acid array in situ fabrication methods and arrays produced using the same
US7206439B2 (en) 2003-04-30 2007-04-17 Agilent Technologies, Inc. Feature locations in array reading
US20040219663A1 (en) 2003-04-30 2004-11-04 Page Robert D. Biopolymer array fabrication using different drop deposition heads
US7269518B2 (en) 2003-04-30 2007-09-11 Agilent Technologies, Inc. Chemical array reading
US6916113B2 (en) 2003-05-16 2005-07-12 Agilent Technologies, Inc. Devices and methods for fluid mixing
WO2004103563A2 (en) 2003-05-20 2004-12-02 Fluidigm Corporation Method and system for microfluidic device and imaging thereof
EP1633889B1 (en) 2003-05-30 2010-09-01 The Board Of Trustees Of The University Of Illinois Gene expression profiles that identify genetically elite ungulate mammals
US7276599B2 (en) 2003-06-02 2007-10-02 Isis Pharmaceuticals, Inc. Oligonucleotide synthesis with alternative solvents
US8133670B2 (en) 2003-06-13 2012-03-13 Cold Spring Harbor Laboratory Method for making populations of defined nucleic acid molecules
US6938476B2 (en) 2003-06-25 2005-09-06 Agilent Technologies, Inc. Apparatus and methods for sensing fluid levels
US7534563B2 (en) 2003-06-30 2009-05-19 Agilent Technologies, Inc. Methods for producing ligand arrays
US20050016851A1 (en) 2003-07-24 2005-01-27 Jensen Klavs F. Microchemical method and apparatus for synthesis and coating of colloidal nanoparticles
US6843281B1 (en) 2003-07-30 2005-01-18 Agilent Techinologies, Inc. Methods and apparatus for introducing liquids into microfluidic chambers
US7353116B2 (en) 2003-07-31 2008-04-01 Agilent Technologies, Inc. Chemical array with test dependent signal reading or processing
US7939310B2 (en) 2003-08-06 2011-05-10 University Of Massachusetts Systems and methods for analyzing nucleic acid sequences
US7028536B2 (en) 2004-06-29 2006-04-18 Nanostream, Inc. Sealing interface for microfluidic device
US7348144B2 (en) 2003-08-13 2008-03-25 Agilent Technologies, Inc. Methods and system for multi-drug treatment discovery
US7229497B2 (en) 2003-08-26 2007-06-12 Massachusetts Institute Of Technology Method of preparing nanocrystals
US7585970B2 (en) 2003-08-30 2009-09-08 Agilent Technologies, Inc. Method of polynucleotide synthesis using modified support
US7427679B2 (en) 2003-08-30 2008-09-23 Agilent Technologies, Inc. Precursors for two-step polynucleotide synthesis
US7417139B2 (en) 2003-08-30 2008-08-26 Agilent Technologies, Inc. Method for polynucleotide synthesis
US7385050B2 (en) 2003-08-30 2008-06-10 Agilent Technologies, Inc. Cleavable linker for polynucleotide synthesis
US7193077B2 (en) 2003-08-30 2007-03-20 Agilent Technologies, Inc. Exocyclic amine triaryl methyl protecting groups in two-step polynucleotide synthesis
US20050049796A1 (en) 2003-09-03 2005-03-03 Webb Peter G. Methods for encoding non-biological information on microarrays
US20120202710A1 (en) 2003-09-09 2012-08-09 Integrigen, Inc. Methods and compositions for generation of germline human antibody genes
EP1687445A4 (en) 2003-09-23 2007-03-28 Atom Sciences Inc Polymeric nucleic acid hybridization probes
US7488607B2 (en) 2003-09-30 2009-02-10 Agilent Technologies, Inc. Electronically readable microarray with electronic addressing function
US7147362B2 (en) 2003-10-15 2006-12-12 Agilent Technologies, Inc. Method of mixing by intermittent centrifugal force
US7075161B2 (en) 2003-10-23 2006-07-11 Agilent Technologies, Inc. Apparatus and method for making a low capacitance artificial nanopore
US20050277125A1 (en) 2003-10-27 2005-12-15 Massachusetts Institute Of Technology High-density reaction chambers and methods of use
US7169560B2 (en) 2003-11-12 2007-01-30 Helicos Biosciences Corporation Short cycle methods for sequencing polynucleotides
US7276338B2 (en) 2003-11-17 2007-10-02 Jacobson Joseph M Nucleotide sequencing via repetitive single molecule hybridization
DE10353887A1 (en) 2003-11-18 2005-06-16 Febit Ag Highly parallel matrix-based DNA synthesizer
US7851192B2 (en) 2004-11-22 2010-12-14 New England Biolabs, Inc. Modified DNA cleavage enzymes and methods for use
US7282705B2 (en) 2003-12-19 2007-10-16 Agilent Technologies, Inc. Microdevice having an annular lining for producing an electrospray emitter
JP2007525571A (en) 2004-01-07 2007-09-06 ソレクサ リミテッド Modified molecular array
US7084180B2 (en) * 2004-01-28 2006-08-01 Velocys, Inc. Fischer-tropsch synthesis using microchannel technology and novel catalyst and microchannel reactor
US7927797B2 (en) 2004-01-28 2011-04-19 454 Life Sciences Corporation Nucleic acid amplification with continuous flow emulsion
US7125488B2 (en) 2004-02-12 2006-10-24 Varian, Inc. Polar-modified bonded phase materials for chromatographic separations
DE602005018166D1 (en) 2004-02-12 2010-01-21 Population Genetics Technologi GENETIC ANALYSIS BY SEQUENCE-SPECIFIC SORTING
WO2005089110A2 (en) 2004-02-27 2005-09-29 President And Fellows Of Harvard College Polynucleotide synthesis
WO2005093092A2 (en) 2004-03-26 2005-10-06 Bayer Healthcare Ag Diagnostics and therapeutics for diseases associated with g-protein coupled receptor 44 (gpr44)
US7875463B2 (en) 2004-03-26 2011-01-25 Agilent Technologies, Inc. Generalized pulse jet ejection head control model
US20050214779A1 (en) 2004-03-29 2005-09-29 Peck Bill J Methods for in situ generation of nucleic acid arrays
US20050214778A1 (en) 2004-03-29 2005-09-29 Peck Bill J Methods for in situ generation of nucleic acid arrays
US8825411B2 (en) 2004-05-04 2014-09-02 Dna Twopointo, Inc. Design, synthesis and assembly of synthetic nucleic acids
US20060003958A1 (en) 2004-05-11 2006-01-05 Melville Mark W Novel polynucleotides related to oligonucleotide arrays to monitor gene expression
PT1773978E (en) 2004-05-19 2014-05-29 Massachusetts Inst Technology Perfused three-dimensional cell/tissue disease models
US7302348B2 (en) 2004-06-02 2007-11-27 Agilent Technologies, Inc. Method and system for quantifying and removing spatial-intensity trends in microarray data
US20060024711A1 (en) 2004-07-02 2006-02-02 Helicos Biosciences Corporation Methods for nucleic acid amplification and sequence determination
ES2372503T3 (en) 2004-07-06 2012-01-20 Bioren, Inc. REVISED MUTAGENESIS TO DEVELOP ALTERED POLYPEPTIDES WITH POTENTIATED PROPERTIES.
EP1774300A4 (en) 2004-07-07 2008-07-02 Home Guardian Llc Instrumented mobility assistance device
US7811753B2 (en) 2004-07-14 2010-10-12 Ibis Biosciences, Inc. Methods for repairing degraded DNA
US20060012793A1 (en) 2004-07-19 2006-01-19 Helicos Biosciences Corporation Apparatus and methods for analyzing samples
US7276720B2 (en) 2004-07-19 2007-10-02 Helicos Biosciences Corporation Apparatus and methods for analyzing samples
US20060019084A1 (en) 2004-07-23 2006-01-26 Pearson Laurence T Monolithic composition and method
US20060024678A1 (en) 2004-07-28 2006-02-02 Helicos Biosciences Corporation Use of single-stranded nucleic acid binding proteins in sequencing
WO2006015789A2 (en) 2004-08-03 2006-02-16 Geneart Ag Method for modulating gene expression by modifying the cpg content
WO2006073504A2 (en) 2004-08-04 2006-07-13 President And Fellows Of Harvard College Wobble sequencing
WO2006018044A1 (en) 2004-08-18 2006-02-23 Agilent Technologies, Inc. Microfluidic assembly with coupled microfluidic devices
US7034290B2 (en) 2004-09-24 2006-04-25 Agilent Technologies, Inc. Target support with pattern recognition sites
US7943046B2 (en) 2004-10-01 2011-05-17 Agilent Technologies, Inc Methods and systems for on-column protein delipidation
US20070122817A1 (en) 2005-02-28 2007-05-31 George Church Methods for assembly of high fidelity synthetic polynucleotides
US20070269870A1 (en) 2004-10-18 2007-11-22 George Church Methods for assembly of high fidelity synthetic polynucleotides
US7141807B2 (en) 2004-10-22 2006-11-28 Agilent Technologies, Inc. Nanowire capillaries for mass spectrometry
US20060110744A1 (en) 2004-11-23 2006-05-25 Sampas Nicolas M Probe design methods and microarrays for comparative genomic hybridization and location analysis
US8380441B2 (en) 2004-11-30 2013-02-19 Agilent Technologies, Inc. Systems for producing chemical array layouts
US11268149B2 (en) 2004-12-08 2022-03-08 Cedars-Sinai Medical Center Diagnosis and treatment of inflammatory bowel disease
US7977119B2 (en) 2004-12-08 2011-07-12 Agilent Technologies, Inc. Chemical arrays and methods of using the same
US7439272B2 (en) 2004-12-20 2008-10-21 Varian, Inc. Ultraporous sol gel monoliths
EP1841788A4 (en) 2004-12-22 2009-01-21 Univ Singapore Novel snake toxin
EP1959012A3 (en) 2004-12-29 2009-12-30 Exiqon A/S Novel oligonucleotide compositions and probe sequences useful for detection and analysis of microRNAs and their target mRNAs
CA2594832A1 (en) 2005-01-13 2006-07-20 Codon Devices, Inc. Compositions and methods for protein design
US20060171855A1 (en) 2005-02-03 2006-08-03 Hongfeng Yin Devices,systems and methods for multi-dimensional separation
WO2006086391A2 (en) 2005-02-07 2006-08-17 Massachusetts Institute Of Technology Electrochemically-degradable layer-by-layer thin films
US7393665B2 (en) 2005-02-10 2008-07-01 Population Genetics Technologies Ltd Methods and compositions for tagging and identifying polynucleotides
JP4641199B2 (en) 2005-02-28 2011-03-02 国立感染症研究所長 Apparatus for designing RNA interference polynucleotide mixture, method for producing RNA interference polynucleotide mixture, and program for designing RNA interference polynucleotide mixture
US20060203236A1 (en) 2005-03-08 2006-09-14 Zhenghua Ji Sample cell
WO2007086890A2 (en) 2005-03-10 2007-08-02 Genemark Inc. Method, apparatus, and system for authentication using labels containing nucleotide seouences
EP1623763A1 (en) 2005-03-11 2006-02-08 Agilent Technologies, Inc. Chip with cleaning cavity
US7618777B2 (en) 2005-03-16 2009-11-17 Agilent Technologies, Inc. Composition and method for array hybridization
US20060219637A1 (en) 2005-03-29 2006-10-05 Killeen Kevin P Devices, systems and methods for liquid chromatography
EP1874792B1 (en) 2005-04-27 2016-04-13 Sigma-Aldrich Co. LLC Activators for oligonucleotide and phosphoramidite synthesis
EP1885880B1 (en) 2005-04-29 2010-07-21 Synthetic Genomics, Inc. Amplification and cloning of single dna molecules using rolling circle amplification
US7572907B2 (en) 2005-04-29 2009-08-11 Agilent Technologies, Inc. Methods and compounds for polynucleotide synthesis
JP5294846B2 (en) 2005-05-12 2013-09-18 ヘンペル エイ/エス Method for fixing crack-resistant epoxy coating film and coating composition suitable for the method
US7396676B2 (en) 2005-05-31 2008-07-08 Agilent Technologies, Inc. Evanescent wave sensor with attached ligand
JP5331476B2 (en) 2005-06-15 2013-10-30 カリダ・ジェノミックス・インコーポレイテッド Single molecule array for genetic and chemical analysis
US7919239B2 (en) 2005-07-01 2011-04-05 Agilent Technologies, Inc. Increasing hybridization efficiencies
US7718365B2 (en) 2005-07-09 2010-05-18 Agilent Technologies, Inc. Microarray analysis of RNA
US8076064B2 (en) 2005-07-09 2011-12-13 Agilent Technologies, Inc. Method of treatment of RNA sample
US20070031857A1 (en) 2005-08-02 2007-02-08 Rubicon Genomics, Inc. Compositions and methods for processing and amplification of DNA, including using multiple enzymes in a single reaction
DE102005037351B3 (en) 2005-08-08 2007-01-11 Geneart Ag In vitro method for directed evolution of proteins, useful e.g. in pharmaceutical development, uses expression system for performing translation, transcription and reverse transcription
MY143596A (en) 2005-08-11 2011-06-15 Synthetic Genomics Inc In vitro recombination method
US7776532B2 (en) 2005-08-11 2010-08-17 Synthetic Genomics, Inc. Method for in vitro recombination
US9404882B2 (en) * 2005-08-11 2016-08-02 New Mexico Tech Research Foundation Method of producing a multi-microchannel, flow-through element and device using same
US7749701B2 (en) 2005-08-11 2010-07-06 Agilent Technologies, Inc. Controlling use of oligonucleotide sequences released from arrays
US7805252B2 (en) 2005-08-16 2010-09-28 Dna Twopointo, Inc. Systems and methods for designing and ordering polynucleotides
WO2007025059A1 (en) 2005-08-26 2007-03-01 Surmodics, Inc. Silane coating compositions, coating systems, and methods
US20070196834A1 (en) 2005-09-09 2007-08-23 Francesco Cerrina Method and system for the generation of large double stranded DNA fragments
WO2007033176A2 (en) 2005-09-14 2007-03-22 Illumina, Inc. Continuous polymer synthesizer
US20100233429A1 (en) 2005-09-16 2010-09-16 Yamatake Corporation Substrate for Biochip, Biochip, Method for Manufacturing Substrate for Biochip and Method for Manufacturing Biochip
US20080308884A1 (en) 2005-10-13 2008-12-18 Silex Microsystems Ab Fabrication of Inlet and Outlet Connections for Microfluidic Chips
US7759471B2 (en) 2005-10-31 2010-07-20 Agilent Technologies, Inc. Monomer compositions for the synthesis of RNA, methods of synthesis, and methods of deprotection
US7368550B2 (en) 2005-10-31 2008-05-06 Agilent Technologies, Inc. Phosphorus protecting groups
US8552174B2 (en) 2005-10-31 2013-10-08 Agilent Technologies, Inc. Solutions, methods, and processes for deprotection of polynucleotides
US8202985B2 (en) 2005-10-31 2012-06-19 Agilent Technologies, Inc. Monomer compositions for the synthesis of polynucleotides, methods of synthesis, and methods of deprotection
GB0522310D0 (en) 2005-11-01 2005-12-07 Solexa Ltd Methods of preparing libraries of template polynucleotides
US7291471B2 (en) 2005-11-21 2007-11-06 Agilent Technologies, Inc. Cleavable oligonucleotide arrays
GB0524069D0 (en) 2005-11-25 2006-01-04 Solexa Ltd Preparation of templates for solid phase amplification
US8137936B2 (en) 2005-11-29 2012-03-20 Macevicz Stephen C Selected amplification of polynucleotides
JP5198284B2 (en) 2005-12-22 2013-05-15 キージーン ナムローゼ フェンノートシャップ An improved strategy for transcript characterization using high-throughput sequencing techniques
EP1989318B1 (en) 2006-01-06 2014-07-30 Agilent Technologies, Inc. Reaction buffer composition for nucleic acid replication with packed dna polymerases
WO2007081386A2 (en) 2006-01-11 2007-07-19 Raindance Technologies, Inc. Microfluidic devices and methods of use
WO2007087310A2 (en) 2006-01-23 2007-08-02 Population Genetics Technologies Ltd. Nucleic acid analysis using sequence tokens
WO2007087377A2 (en) 2006-01-25 2007-08-02 Massachusetts Institute Of Technology Photoelectrochemical synthesis of high density combinatorial polymer arrays
WO2008057127A2 (en) 2006-02-06 2008-05-15 Massachusetts Institute Of Technology Self-assembly of macromolecules on multilayered polymer surfaces
US8809876B2 (en) 2006-02-14 2014-08-19 Massachusetts Institute Of Technology Absorbing film
US7807356B2 (en) 2006-03-09 2010-10-05 Agilent Technologies, Inc. Labeled nucleotide composition
TW200806317A (en) 2006-03-20 2008-02-01 Wyeth Corp Methods for reducing protein aggregation
US7855281B2 (en) 2006-03-23 2010-12-21 Agilent Technologies, Inc. Cleavable thiocarbonate linkers for polynucleotide synthesis
US7572908B2 (en) 2006-03-23 2009-08-11 Agilent Technologies, Inc. Cleavable linkers for polynucleotides
US20070231800A1 (en) 2006-03-28 2007-10-04 Agilent Technologies, Inc. Determination of methylated DNA
EP3373174A1 (en) 2006-03-31 2018-09-12 Illumina, Inc. Systems and devices for sequence by synthesis analysis
US8058055B2 (en) 2006-04-07 2011-11-15 Agilent Technologies, Inc. High resolution chromosomal mapping
US20070238106A1 (en) 2006-04-07 2007-10-11 Agilent Technologies, Inc. Systems and methods of determining alleles and/or copy numbers
US20070238108A1 (en) 2006-04-07 2007-10-11 Agilent Technologies, Inc. Validation of comparative genomic hybridization
KR20090029184A (en) 2006-04-07 2009-03-20 더 가브먼트 오브 더 유나이티드 스테이츠 오브 아메리카, 리프리젠티드 바이 더 세크러테리, 디파트먼트 오브 헬쓰 앤드 휴먼 서비씨즈 Antibody compositions and methods for treatment of neoplastic disease
US20070238104A1 (en) 2006-04-07 2007-10-11 Agilent Technologies, Inc. Competitive oligonucleotides
JP5654749B2 (en) 2006-04-11 2015-01-14 ニユー・イングランド・バイオレイブス・インコーポレイテツド Nucleic acid repair for improved amplification
JP2009538123A (en) 2006-04-19 2009-11-05 アプライド バイオシステムズ, エルエルシー Reagents, methods and libraries for gel-free bead-based sequencing
US8383338B2 (en) 2006-04-24 2013-02-26 Roche Nimblegen, Inc. Methods and systems for uniform enrichment of genomic regions
US20070259344A1 (en) 2006-05-03 2007-11-08 Agilent Technologies, Inc. Compound probes and methods of increasing the effective probe densities of arrays
US20070259347A1 (en) 2006-05-03 2007-11-08 Agilent Technologies, Inc. Methods of increasing the effective probe densities of arrays
US20070259346A1 (en) 2006-05-03 2007-11-08 Agilent Technologies, Inc. Analysis of arrays
US20070259345A1 (en) 2006-05-03 2007-11-08 Agilent Technologies, Inc. Target determination using compound probes
WO2007136834A2 (en) 2006-05-19 2007-11-29 Codon Devices, Inc. Combined extension and ligation for nucleic acid assembly
WO2007137242A2 (en) 2006-05-19 2007-11-29 Massachusetts Institute Of Technology Microfluidic-based gene synthesis
WO2008054543A2 (en) 2006-05-20 2008-05-08 Codon Devices, Inc. Oligonucleotides for multiplex nucleic acid assembly
EP2035984A4 (en) 2006-06-19 2010-03-31 Yeda Res & Dev Programmable iterated elongation: a method for manufacturing synthetic genes and combinatorial dna and protein libraries
AT503902B1 (en) 2006-07-05 2008-06-15 F Star Biotech Forsch & Entw METHOD FOR MANIPULATING IMMUNE LOBULINS
AT503861B1 (en) 2006-07-05 2008-06-15 F Star Biotech Forsch & Entw METHOD FOR MANIPULATING T-CELL RECEPTORS
US20080193772A1 (en) 2006-07-07 2008-08-14 Bio-Rad Laboratories, Inc Mass spectrometry probes having hydrophobic coatiings
US7524942B2 (en) 2006-07-31 2009-04-28 Agilent Technologies, Inc. Labeled nucleotide composition
US7572585B2 (en) 2006-07-31 2009-08-11 Agilent Technologies, Inc. Enzymatic labeling of RNA
WO2008015396A2 (en) 2006-07-31 2008-02-07 Solexa Limited Method of library preparation avoiding the formation of adaptor dimers
SI2056845T1 (en) 2006-08-08 2018-02-28 Rheinische Friedrich-Wilhelms-Universitaet Bonn Structure and use of 5' phosphate oligonucleotides
DE102006039479A1 (en) 2006-08-23 2008-03-06 Febit Biotech Gmbh Programmable oligonucleotide synthesis
WO2008023179A2 (en) 2006-08-24 2008-02-28 Solexa Limited Method for retaining even coverage of short insert libraries
US8053191B2 (en) 2006-08-31 2011-11-08 Westend Asset Clearinghouse Company, Llc Iterative nucleic acid assembly using activation of vector-encoded traits
US8415138B2 (en) 2006-08-31 2013-04-09 Agilent Technologies, Inc. Apparatuses and methods for oligonucleotide preparation
US8097711B2 (en) 2006-09-02 2012-01-17 Agilent Technologies, Inc. Thioether substituted aryl carbonate protecting groups
US20080311628A1 (en) 2006-10-03 2008-12-18 Ghc Technologies, Inc. Methods and compositions for rapid amplification and capture of nucleic acid sequences
WO2008045380A2 (en) 2006-10-04 2008-04-17 Codon Devices, Inc. Nucleic acid libraries and their design and assembly
US20080085511A1 (en) 2006-10-05 2008-04-10 Peck Bill J Preparation of biopolymer arrays
JP2008097189A (en) 2006-10-10 2008-04-24 National Institute Of Advanced Industrial & Technology Method for judging transfer object specificity or gene specificity of base sequence fragment
US20080085514A1 (en) 2006-10-10 2008-04-10 Peck Bill J Methods and devices for array synthesis
US7867782B2 (en) 2006-10-19 2011-01-11 Agilent Technologies, Inc. Nanoscale moiety placement methods
US7999087B2 (en) 2006-11-15 2011-08-16 Agilent Technologies, Inc. 2′-silyl containing thiocarbonate protecting groups for RNA synthesis
WO2008063135A1 (en) 2006-11-24 2008-05-29 Agency For Science, Technology And Research Apparatus for processing a sample in a liquid droplet and method of using the same
WO2008063134A1 (en) 2006-11-24 2008-05-29 Agency For Science, Technology And Research Method of producing a pattern of discriminative wettability
US8242258B2 (en) 2006-12-03 2012-08-14 Agilent Technologies, Inc. Protecting groups for RNA synthesis
US7989396B2 (en) 2006-12-05 2011-08-02 The Board Of Trustees Of The Leland Stanford Junior University Biomolecule immobilization on biosensors
JP2010511397A (en) 2006-12-05 2010-04-15 アブリンクス エン.ヴェー. Peptides that can bind to serum proteins
US7862999B2 (en) 2007-01-17 2011-01-04 Affymetrix, Inc. Multiplex targeted amplification using flap nuclease
US8314220B2 (en) 2007-01-26 2012-11-20 Agilent Technologies, Inc. Methods compositions, and kits for detection of microRNA
US20080182296A1 (en) 2007-01-31 2008-07-31 Chanda Pranab K Pcr-directed gene synthesis from large number of overlapping oligodeoxyribonucleotides
KR100827449B1 (en) 2007-02-07 2008-05-07 삼성전자주식회사 Photolabile compound, oligomer probe array and substrate for oligomer probe array with the same and the method of fabricating the same
WO2008115632A2 (en) 2007-02-09 2008-09-25 The Regents Of The University Of California Method for recombining dna sequences and compositions related thereto
EP2126105A4 (en) 2007-02-20 2010-11-03 Anaptysbio Inc Somatic hypermutation systems
WO2008109176A2 (en) 2007-03-07 2008-09-12 President And Fellows Of Harvard College Assays and other reactions involving droplets
US7651762B2 (en) 2007-03-13 2010-01-26 Varian, Inc. Methods and devices using a shrinkable support for porous monolithic materials
EP3798317B1 (en) 2007-04-04 2024-01-03 The Regents of the University of California Compositions, devices, systems, and methods for using a nanopore
EP2160396B1 (en) 2007-05-10 2018-11-21 Agilent Technologies, Inc. Thiocarbon-protecting groups for rna synthesis
WO2008148392A1 (en) 2007-06-04 2008-12-11 In Situ Rcp A/S Enzyme activity assay using rolling circle amplification
US20090023190A1 (en) 2007-06-20 2009-01-22 Kai Qin Lao Sequence amplification with loopable primers
US20080318334A1 (en) 2007-06-20 2008-12-25 Robotti Karla M Microfluidic devices comprising fluid flow paths having a monolithic chromatographic material
US8194244B2 (en) 2007-06-29 2012-06-05 Intel Corporation Solution sample plate with wells designed for improved Raman scattering signal detection efficiency
US7659069B2 (en) 2007-08-31 2010-02-09 Agilent Technologies, Inc. Binary signaling assay using a split-polymerase
US8685642B2 (en) 2007-07-30 2014-04-01 Agilent Technologies, Inc. Allele-specific copy number measurement using single nucleotide polymorphism and DNA arrays
US7979215B2 (en) 2007-07-30 2011-07-12 Agilent Technologies, Inc. Methods and systems for evaluating CGH candidate probe nucleic acid sequences
US20090036664A1 (en) 2007-07-31 2009-02-05 Brian Jon Peter Complex oligonucleotide primer mix
WO2009020435A1 (en) 2007-08-07 2009-02-12 Agency For Science, Technology And Research Integrated microfluidic device for gene synthesis
US9440231B2 (en) 2007-08-14 2016-09-13 Fluidigm Corporation Polymer microfluidic biochip fabrication
US20110126929A1 (en) 2007-08-15 2011-06-02 Massachusetts Institute Of Technology Microstructures For Fluidic Ballasting and Flow Control
US20090053704A1 (en) 2007-08-24 2009-02-26 Natalia Novoradovskaya Stabilization of nucleic acids on solid supports
US9598737B2 (en) 2012-05-09 2017-03-21 Longhorn Vaccines And Diagnostics, Llc Next generation genomic sequencing methods
US8877688B2 (en) 2007-09-14 2014-11-04 Adimab, Llc Rationally designed, synthetic antibody libraries and uses therefor
CA2699518A1 (en) 2007-09-17 2009-03-26 Twof, Inc. Supramolecular nanostamping printing device
US7790387B2 (en) 2007-09-24 2010-09-07 Agilent Technologies, Inc. Thiocarbonate linkers for polynucleotides
WO2009045344A2 (en) 2007-09-28 2009-04-09 Pacific Biosciences Of California, Inc. Error-free amplification of dna for clonal sequencing
EP2053132A1 (en) 2007-10-23 2009-04-29 Roche Diagnostics GmbH Enrichment and sequence analysis of geomic regions
WO2009070665A1 (en) 2007-11-27 2009-06-04 Massachusetts Institute Of Technology Near field detector for integrated surface plasmon resonance biosensor applications
WO2009076580A2 (en) 2007-12-12 2009-06-18 Thomas Jefferson University Compositions and methods for the treatment and prevention of cardiovascular diseases
US9286439B2 (en) 2007-12-17 2016-03-15 Yeda Research And Development Co Ltd System and method for editing and manipulating DNA
US9540637B2 (en) 2008-01-09 2017-01-10 Life Technologies Corporation Nucleic acid adaptors and uses thereof
CN102016068A (en) 2008-01-09 2011-04-13 生命科技公司 Method of making a paired tag library for nucleic acid sequencing
US7682809B2 (en) 2008-01-11 2010-03-23 Agilent Technologies, Inc. Direct ATP release sequencing
WO2009092564A2 (en) 2008-01-23 2009-07-30 Roche Diagnostics Gmbh Integrated instrument performing synthesis and amplification
WO2009131724A2 (en) 2008-01-24 2009-10-29 Massachusetts Institute Of Technology Insulated nanogap devices and methods of use thereof
WO2009097368A2 (en) 2008-01-28 2009-08-06 Complete Genomics, Inc. Methods and compositions for efficient base calling in sequencing reactions
US20090194483A1 (en) 2008-01-31 2009-08-06 Robotti Karla M Microfluidic device having monolithic separation medium and method of use
DK2255013T3 (en) 2008-02-15 2016-09-12 Synthetic Genomics Inc Methods for in vitro joining and combinatorial assembly of nucleic acid molecules.
WO2009113709A1 (en) 2008-03-11 2009-09-17 国立大学法人東京大学 Method of preparing dna fragment having sticky end
US20090230044A1 (en) 2008-03-13 2009-09-17 Agilent Technologies, Inc. Microfluid Chip Cleaning
US20090238722A1 (en) 2008-03-18 2009-09-24 Agilent Technologies, Inc. Pressure-Reinforced Fluidic Chip
EP2286217B1 (en) 2008-03-31 2015-02-18 Pacific Biosciences of California, Inc. Single molecule loading methods and compositions
US20090246788A1 (en) 2008-04-01 2009-10-01 Roche Nimblegen, Inc. Methods and Assays for Capture of Nucleic Acids
CA2720587A1 (en) 2008-04-09 2009-10-15 Francis Barany Coferons and methods of making and using them
US8911948B2 (en) 2008-04-30 2014-12-16 Integrated Dna Technologies, Inc. RNase H-based assays utilizing modified RNA monomers
EP2113255A1 (en) 2008-05-02 2009-11-04 f-star Biotechnologische Forschungs- und Entwicklungsges.m.b.H. Cytotoxic immunoglobulin
JP4582224B2 (en) 2008-05-02 2010-11-17 ソニー株式会社 Microbead manufacturing method and microbead
EP3733871A1 (en) 2008-05-27 2020-11-04 Dako Denmark A/S Compositions and methods for detection of chromosomal aberrations with novel hybridization buffers
JP5654986B2 (en) 2008-06-30 2015-01-14 モーフオテク・インコーポレーテツド Anti-GD2 antibodies and related methods and uses
GB2461546B (en) 2008-07-02 2010-07-07 Argen X Bv Antigen binding polypeptides
JP4667490B2 (en) 2008-07-09 2011-04-13 三菱電機株式会社 Cooker
WO2010014903A1 (en) 2008-07-31 2010-02-04 Massachusetts Institute Of Technology Multiplexed olfactory receptor-based microsurface plasmon polariton detector
US20100069250A1 (en) 2008-08-16 2010-03-18 The Board Of Trustees Of The Leland Stanford Junior University Digital PCR Calibration for High Throughput Sequencing
US8703489B2 (en) 2008-08-22 2014-04-22 Sangamo Biosciences, Inc. Methods and compositions for targeted single-stranded cleavage and targeted integration
WO2010025310A2 (en) 2008-08-27 2010-03-04 Westend Asset Clearinghouse Company, Llc Methods and devices for high fidelity polynucleotide synthesis
US8034917B2 (en) 2008-08-28 2011-10-11 Agilent Technologies, Inc. Primer-directed chromosome painting
EP2334692B1 (en) 2008-09-05 2016-04-13 The Royal Institution for the Advancement of Learning/McGill University Rna monomers containing o-acetal levulinyl ester groups and their use in rna microarrays
US8586310B2 (en) 2008-09-05 2013-11-19 Washington University Method for multiplexed nucleic acid patch polymerase chain reaction
JP2012501658A (en) 2008-09-05 2012-01-26 ライフ テクノロジーズ コーポレーション Methods and systems for nucleic acid sequencing validation, calibration, and standardization
JP5714490B2 (en) 2008-09-06 2015-05-07 ケムジーンズ コーポレーション Methods for preparing oligonucleotides by RNA synthesis, phosphoramidites for synthetic RNA in the reverse direction, and bond formation in the 5 'to 3' direction for synthesis of RNA oligomers
US8541569B2 (en) 2008-09-06 2013-09-24 Chemgenes Corporation Phosphoramidites for synthetic RNA in the reverse direction, efficient RNA synthesis and convenient introduction of 3'-end ligands, chromophores and modifications of synthetic RNA
WO2010030776A1 (en) 2008-09-10 2010-03-18 Genscript Corporation Homologous recombination-based dna cloning methods and compositions
US20100076183A1 (en) 2008-09-22 2010-03-25 Dellinger Douglas J Protected monomer and method of final deprotection for rna synthesis
US8213015B2 (en) 2008-09-25 2012-07-03 Agilent Technologies, Inc. Integrated flow cell with semiconductor oxide tubing
TW201028432A (en) 2008-09-30 2010-08-01 Abbott Lab Improved antibody libraries
US20100090341A1 (en) 2008-10-14 2010-04-15 Molecular Imprints, Inc. Nano-patterned active layers formed by nano-imprint lithography
US20100301398A1 (en) 2009-05-29 2010-12-02 Ion Torrent Systems Incorporated Methods and apparatus for measuring analytes
US9080211B2 (en) 2008-10-24 2015-07-14 Epicentre Technologies Corporation Transposon end compositions and methods for modifying nucleic acids
US8357489B2 (en) 2008-11-13 2013-01-22 The Board Of Trustees Of The Leland Stanford Junior University Methods for detecting hepatocellular carcinoma
WO2010062960A2 (en) 2008-11-26 2010-06-03 Cedars-Sinai Medical Center METHODS OF DETERMINING RESPONSIVENESS TO ANTI-TNFα THERAPY IN INFLAMMATORY BOWEL DISEASE
KR101881596B1 (en) 2008-12-02 2018-07-24 웨이브 라이프 사이언시스 재팬 인코포레이티드 Method for the synthesis of phosphorous atom modified nucleic acids
US8963262B2 (en) 2009-08-07 2015-02-24 Massachusettes Institute Of Technology Method and apparatus for forming MEMS device
JO3382B1 (en) 2008-12-23 2019-03-13 Amgen Inc Human cgrp receptor binding antibodies
TW201104253A (en) 2008-12-31 2011-02-01 Nat Health Research Institutes Microarray chip and method of fabricating for the same
AU2010210083C1 (en) 2009-02-09 2015-11-26 Helmholtz Zentrum Muenchen Deutsches Forschungszentrum Fuer Gesundheit Und Umwelt (Gmbh) Repertoire of allo-restricted peptide-specific T cell receptor sequences and use thereof
US8569046B2 (en) 2009-02-20 2013-10-29 Massachusetts Institute Of Technology Microarray with microchannels
WO2010094772A1 (en) 2009-02-20 2010-08-26 Febit Holding Gmbh Synthesis of sequence-verified nucleic acids
EP2406399B1 (en) 2009-03-09 2018-02-14 Bioatla, LLC Mirac proteins
WO2010115122A2 (en) 2009-04-03 2010-10-07 Illumina, Inc. Generation of uniform fragments of nucleic acids using patterned substrates
US7862716B2 (en) 2009-04-13 2011-01-04 Sielc Technologies Corporation HPLC schematic with integrated sample cleaning system
CN102802798B (en) 2009-04-29 2015-04-08 锡克拜控股有限公司 Method and apparatus for depositing a biological fluid onto a substrate
WO2010127186A1 (en) 2009-04-30 2010-11-04 Prognosys Biosciences, Inc. Nucleic acid constructs and methods of use
EP2248914A1 (en) 2009-05-05 2010-11-10 Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V. The use of class IIB restriction endonucleases in 2nd generation sequencing applications
US9309557B2 (en) 2010-12-17 2016-04-12 Life Technologies Corporation Nucleic acid amplification
US20100292102A1 (en) 2009-05-14 2010-11-18 Ali Nouri System and Method For Preventing Synthesis of Dangerous Biological Sequences
US20100300882A1 (en) * 2009-05-26 2010-12-02 General Electric Company Devices and methods for in-line sample preparation of materials
WO2010141433A2 (en) 2009-06-02 2010-12-09 The Regents Of The University Of California Virus discovery by sequencing and assembly of virus-derived sirnas, mirnas, pirnas
WO2010141249A2 (en) 2009-06-02 2010-12-09 Merck Sharp & Dohme Corp. Generation, characterization and uses thereof of anti-notch3 antibodies
US8309710B2 (en) 2009-06-29 2012-11-13 Agilent Technologies, Inc. Use of N-alkyl imidazole for sulfurization of oligonucleotides with an acetyl disulfide
US8642755B2 (en) 2009-06-30 2014-02-04 Agilent Technologies, Inc. Use of thioacetic acid derivatives in the sulfurization of oligonucleotides with phenylacetyl disulfide
GB0912909D0 (en) 2009-07-23 2009-08-26 Olink Genomics Ab Probes for specific analysis of nucleic acids
US8329208B2 (en) 2009-07-28 2012-12-11 Methylation Sciences International Srl Pharmacokinetics of S-adenosylmethionine formulations
SG177485A1 (en) 2009-07-30 2012-02-28 Hoffmann La Roche A set of oligonucleotide probes as well as methods and uses related thereto
JP5734978B2 (en) 2009-08-19 2015-06-17 メルク パテント ゲゼルシャフト ミット ベシュレンクテル ハフツングMerck Patent Gesellschaft mit beschraenkter Haftung Antibodies for integrin complex detection in FFPE materials
EP2467479B1 (en) 2009-08-20 2016-01-06 Population Genetics Technologies Ltd Compositions and methods for intramolecular nucleic acid rearrangement
US8476598B1 (en) 2009-08-31 2013-07-02 Sionyx, Inc. Electromagnetic radiation imaging devices and associated methods
US20110082055A1 (en) 2009-09-18 2011-04-07 Codexis, Inc. Reduced codon mutagenesis
US20120184724A1 (en) 2009-09-22 2012-07-19 Agilent Technologies, Inc. Protected monomers and methods of deprotection for rna synthesis
WO2011038241A1 (en) 2009-09-25 2011-03-31 President And Fellows Of Harvard College Nucleic acid amplification and sequencing by synthesis with fluorogenic nucleotides
US8975019B2 (en) 2009-10-19 2015-03-10 University Of Massachusetts Deducing exon connectivity by RNA-templated DNA ligation/sequencing
US8389689B2 (en) 2009-10-28 2013-03-05 Janssen Biotech, Inc. Anti-GLP-1R antibodies and their uses
US20120315670A1 (en) 2009-11-02 2012-12-13 Gen9, Inc. Compositions and Methods for the Regulation of Multiple Genes of Interest in a Cell
WO2011056872A2 (en) 2009-11-03 2011-05-12 Gen9, Inc. Methods and microfluidic devices for the manipulation of droplets in high fidelity polynucleotide assembly
US20110114549A1 (en) 2009-11-13 2011-05-19 Agilent Technolgies, Inc. Microfluidic device comprising separation columns
EP2504449B1 (en) 2009-11-25 2016-03-23 Gen9, Inc. Methods and apparatuses for chip-based dna error reduction
WO2011066185A1 (en) 2009-11-25 2011-06-03 Gen9, Inc. Microfluidic devices and methods for gene synthesis
US8500979B2 (en) 2009-12-31 2013-08-06 Intel Corporation Nanogap chemical and biochemical sensors
US9217144B2 (en) 2010-01-07 2015-12-22 Gen9, Inc. Assembly of high fidelity polynucleotides
US9758817B2 (en) 2010-01-13 2017-09-12 Agilent Technologies, Inc. Method for identifying a nucleic acid in a sample
KR101230350B1 (en) 2010-01-27 2013-02-06 주식회사 엘지화학 Battery Pack of Excellent Structural Stability
KR20110094878A (en) 2010-02-18 2011-08-24 삼성전자주식회사 Composition of forming an oligomer array and methods of forming an oligomer array
US20120027786A1 (en) 2010-02-23 2012-02-02 Massachusetts Institute Of Technology Genetically programmable pathogen sense and destroy
GB201003036D0 (en) 2010-02-23 2010-04-07 Fermentas Uab Restriction endonucleases and their applications
US8716467B2 (en) 2010-03-03 2014-05-06 Gen9, Inc. Methods and devices for nucleic acid synthesis
JP2013520989A (en) 2010-03-05 2013-06-10 シンセティック ジェノミクス インコーポレーテッド Methods for genome cloning and manipulation
US20130085472A1 (en) 2010-03-09 2013-04-04 Toxcure, LLC Microneedle nasal delivery device
CN103725703B (en) 2010-04-09 2016-02-10 美国天主教大学 For the preparation of method and the packaging machine of packaging machine
WO2011130223A1 (en) 2010-04-12 2011-10-20 Mayo Foundation For Medical Education And Research System and method for alleviating freezing gait and gait hypokinesia in users with extrapyramidal disorders
WO2011143556A1 (en) 2010-05-13 2011-11-17 Gen9, Inc. Methods for nucleotide sequencing and high fidelity polynucleotide synthesis
KR101339064B1 (en) 2010-05-28 2014-01-06 한양대학교 에리카산학협력단 Mehthod of decoding codes using a bio-chip and mehtod of determining whether a bar-code is forged using the same
WO2011150168A1 (en) 2010-05-28 2011-12-01 Gen9, Inc. Methods and devices for in situ nucleic acid synthesis
GB2481425A (en) 2010-06-23 2011-12-28 Iti Scotland Ltd Method and device for assembling polynucleic acid sequences
CA2805320A1 (en) 2010-07-28 2012-02-02 Immunocore Ltd T cell receptors
DK2623613T3 (en) 2010-09-21 2016-10-03 Population Genetics Tech Ltd Increasing the reliability of the allele-indications by molecular counting
US8715933B2 (en) 2010-09-27 2014-05-06 Nabsys, Inc. Assay methods using nicking endonucleases
WO2012045001A2 (en) 2010-09-30 2012-04-05 Vanderbilt University Influenza virus antibodies and immunogens and uses therefor
US9689012B2 (en) 2010-10-12 2017-06-27 Cornell University Method of dual-adapter recombination for efficient concatenation of multiple DNA fragments in shuffled or specified arrangements
WO2012154201A1 (en) 2010-10-22 2012-11-15 President And Fellows Of Harvard College Orthogonal amplification and assembly of nucleic acid sequences
EP2635679B1 (en) 2010-11-05 2017-04-19 Illumina, Inc. Linking sequence reads using paired code tags
US10457935B2 (en) 2010-11-12 2019-10-29 Gen9, Inc. Protein arrays and methods of using and making the same
IL280133B2 (en) 2010-11-12 2023-04-01 Gen9 Inc Methods and devices for nucleic acids synthesis
EP3564392B1 (en) 2010-12-17 2021-11-24 Life Technologies Corporation Methods for nucleic acid amplification
WO2012092260A1 (en) 2010-12-27 2012-07-05 Ibis Biosciences, Inc. Compositions and methods for producing single-stranded circular dna
US20120164633A1 (en) 2010-12-27 2012-06-28 Ibis Biosciences, Inc. Digital droplet sequencing
MX344595B (en) 2010-12-31 2016-12-20 Bioatla Llc Express humanization of antibodies.
CN104673670A (en) 2011-03-30 2015-06-03 独立行政法人国立长寿医疗研究中心 Membrane-separation-type Culture Device, Membrane-separation-type Culture Kit, Stem Cell Separation Method Using Same, And Separation Membrane
US10131903B2 (en) 2011-04-01 2018-11-20 The Regents Of The University Of California Microfluidic platform for synthetic biology applications
US9384920B1 (en) 2011-04-04 2016-07-05 Eric J. Bakulich Locking knob
WO2012149171A1 (en) 2011-04-27 2012-11-01 The Regents Of The University Of California Designing padlock probes for targeted genomic sequencing
US8722585B2 (en) 2011-05-08 2014-05-13 Yan Wang Methods of making di-tagged DNA libraries from DNA or RNA using double-tagged oligonucleotides
SG10201605049QA (en) 2011-05-20 2016-07-28 Fluidigm Corp Nucleic acid encoding reactions
WO2012167328A1 (en) 2011-06-10 2012-12-13 Bright Devices Group Pty Ltd Freezing of gait cue apparatus
US9752176B2 (en) 2011-06-15 2017-09-05 Ginkgo Bioworks, Inc. Methods for preparative in vitro cloning
PT2723764T (en) 2011-06-21 2018-03-09 Univ Leland Stanford Junior Binding domains directed against gpcr: g protein complexes and uses derived thereof
US9487824B2 (en) 2011-06-28 2016-11-08 Igor Kutyavin Methods and compositions for enrichment of nucleic acids in mixtures of highly homologous sequences
US20130045483A1 (en) 2011-07-01 2013-02-21 Whitehead Institute For Biomedical Research Yeast cells expressing amyloid beta and uses therefor
WO2013019361A1 (en) 2011-07-07 2013-02-07 Life Technologies Corporation Sequencing methods
US20130017978A1 (en) 2011-07-11 2013-01-17 Finnzymes Oy Methods and transposon nucleic acids for generating a dna library
WO2013010062A2 (en) 2011-07-14 2013-01-17 Life Technologies Corporation Nucleic acid complexity reduction
ES2737957T3 (en) 2011-08-26 2020-01-17 Gen9 Inc Compositions and methods for high fidelity nucleic acid assembly
US20150203839A1 (en) 2011-08-26 2015-07-23 Gen9, Inc. Compositions and Methods for High Fidelity Assembly of Nucleic Acids
CN103907117B (en) 2011-09-01 2019-03-29 基因组编译器公司 System and method for polynucleotide constructs design
EP2753714B1 (en) 2011-09-06 2017-04-12 Gen-Probe Incorporated Circularized templates for sequencing
US8840981B2 (en) 2011-09-09 2014-09-23 Eastman Kodak Company Microfluidic device with multilayer coating
US20130109596A1 (en) 2011-09-26 2013-05-02 Life Technologies Corporation High efficiency, small volume nucleic acid synthesis
US20130091126A1 (en) 2011-10-11 2013-04-11 Life Technologies Corporation Systems and methods for analysis and interpretation of nucleic acid sequence data
GB2497838A (en) 2011-10-19 2013-06-26 Nugen Technologies Inc Compositions and methods for directional nucleic acid amplification and sequencing
US8987174B2 (en) 2011-10-28 2015-03-24 Prognosys Biosciences, Inc. Methods for manufacturing molecular arrays
US8815782B2 (en) 2011-11-11 2014-08-26 Agilent Technologies, Inc. Use of DNAzymes for analysis of an RNA sample
EP2599785A1 (en) 2011-11-30 2013-06-05 Agilent Technologies, Inc. Novel methods for the synthesis and purification of oligomers
US20130137173A1 (en) 2011-11-30 2013-05-30 Feng Zhang Nucleotide-specific recognition sequences for designer tal effectors
US8450107B1 (en) 2011-11-30 2013-05-28 The Broad Institute Inc. Nucleotide-specific recognition sequences for designer TAL effectors
WO2013096692A1 (en) 2011-12-21 2013-06-27 Illumina, Inc. Apparatus and methods for kinetic analysis and determination of nucleic acid sequences
WO2013093693A1 (en) 2011-12-22 2013-06-27 Rinat Neuroscience Corp. Staphylococcus aureus specific antibodies and uses thereof
US9593375B2 (en) 2011-12-30 2017-03-14 Quest Diagnostics Investments Incorporated Nucleic acid analysis using emulsion PCR
US20130217071A1 (en) 2011-12-30 2013-08-22 Luz Montesclaros Methods and compositions for performing nucleic acid amplification reactions
DK2809795T3 (en) 2012-02-01 2019-12-16 Sgi Dna Inc MATERIALS AND PROCEDURES FOR SYNTHESIS OF ERROR-MINIMIZED NUCLEIC ACID MOLECULES
ES2776673T3 (en) 2012-02-27 2020-07-31 Univ North Carolina Chapel Hill Methods and uses for molecular tags
WO2013128281A1 (en) 2012-02-28 2013-09-06 Population Genetics Technologies Ltd Method for attaching a counter sequence to a nucleic acid sample
EP3121194A1 (en) 2012-03-14 2017-01-25 Innovative Targeting Solutions Inc. Generating targeted sequence diversity in fusion proteins
US9150853B2 (en) 2012-03-21 2015-10-06 Gen9, Inc. Methods for screening proteins using DNA encoded chemical libraries as templates for enzyme catalysis
CN104168865B (en) 2012-03-28 2016-10-26 凯希特许有限公司 Assist the depressurized system separated with clinical parts of electronics, apply part and method
US9732384B2 (en) 2012-04-02 2017-08-15 Lux Bio Group, Inc. Apparatus and method for molecular separation, purification, and sensing
WO2013154770A1 (en) 2012-04-10 2013-10-17 The Trustees Of Princeton University Ultra-sensitive sensor
US20150353921A9 (en) 2012-04-16 2015-12-10 Jingdong Tian Method of on-chip nucleic acid molecule synthesis
US20130281308A1 (en) 2012-04-24 2013-10-24 Gen9, Inc. Methods for sorting nucleic acids and preparative in vitro cloning
CN104603286B (en) 2012-04-24 2020-07-31 Gen9股份有限公司 Method for sorting nucleic acids and multiplex preparations in vitro cloning
JP6267689B2 (en) 2012-05-10 2018-01-24 バイオアトラ、エルエルシー Multispecific monoclonal antibody
WO2013177220A1 (en) 2012-05-21 2013-11-28 The Scripps Research Institute Methods of sample preparation
ES2698609T3 (en) 2012-06-01 2019-02-05 European Molecular Biology Laboratory High capacity storage of digital information in DNA
US10308979B2 (en) 2012-06-01 2019-06-04 Agilent Technologies, Inc. Target enrichment and labeling for multi-kilobase DNA
US9102936B2 (en) 2012-06-11 2015-08-11 Agilent Technologies, Inc. Method of adaptor-dimer subtraction using a CRISPR CAS6 protein
JP2015521472A (en) 2012-06-14 2015-07-30 フレッド ハチンソン キャンサー リサーチ センター Compositions and methods for sensitive mutation detection in nucleic acid molecules
LT2864531T (en) 2012-06-25 2019-03-12 Gen9, Inc. Methods for nucleic acid assembly and high throughput sequencing
AU2013286635B2 (en) 2012-07-03 2018-11-08 Foundation Medicine, Inc. Tm-enhanced blocking oligonucleotides and baits for improved target enrichment and reduced off-target selection
US9255245B2 (en) 2012-07-03 2016-02-09 Agilent Technologies, Inc. Sample probes and methods for sampling intracellular material
US20140038240A1 (en) 2012-07-10 2014-02-06 Pivot Bio, Inc. Methods for multipart, modular and scarless assembly of dna molecules
US9073962B2 (en) 2012-07-12 2015-07-07 Massachusetts Institute Of Technology Methods of serial assembly of DNA bricks into larger structures
JP6239813B2 (en) 2012-07-18 2017-11-29 株式会社Screenセミコンダクターソリューションズ Substrate processing apparatus and substrate processing method
WO2014014991A2 (en) 2012-07-19 2014-01-23 President And Fellows Of Harvard College Methods of storing information using nucleic acids
MX2015000979A (en) 2012-07-27 2015-11-23 Univ Illinois Engineering t-cell receptors.
WO2014021938A1 (en) 2012-08-02 2014-02-06 The Board Of Trustees Of The Leland Stanford Junior University Methods and apparatus for nucleic acid synthesis using oligo-templated polymerization
WO2014028895A1 (en) 2012-08-16 2014-02-20 Synthetic Genomics, Inc. Digital to biological converter
EP2966088B1 (en) 2012-08-31 2019-10-16 The Scripps Research Institute Antibodies that modulate eukaryotic cells
US9328376B2 (en) 2012-09-05 2016-05-03 Bio-Rad Laboratories, Inc. Systems and methods for stabilizing droplets
US9133510B2 (en) 2012-10-15 2015-09-15 Life Technologies Corporation Compositions, methods, systems and kits for target nucleic acid enrichment
KR20140048733A (en) 2012-10-16 2014-04-24 삼성전자주식회사 Multiwell plate and method for analyzing target material using the same
WO2014066179A1 (en) 2012-10-24 2014-05-01 Clontech Laboratories, Inc. Template switch-based methods for producing a product nucleic acid
US11439594B2 (en) 2012-12-04 2022-09-13 Phosphorex, Inc. Microparticles and nanoparticles having negative surface charges
US10662424B2 (en) 2012-12-06 2020-05-26 Agilent Technologies, Inc. Molecular fabrication
US10072260B2 (en) 2012-12-06 2018-09-11 Agilent Technologies, Inc. Target enrichment of randomly sheared genomic DNA fragments
US9932576B2 (en) 2012-12-10 2018-04-03 Resolution Bioscience, Inc. Methods for targeted genomic analysis
WO2014092886A2 (en) 2012-12-10 2014-06-19 Agilent Technologies, Inc. Pairing code directed assembly
US20140310830A1 (en) 2012-12-12 2014-10-16 Feng Zhang CRISPR-Cas Nickase Systems, Methods And Compositions For Sequence Manipulation in Eukaryotes
EP2962110A4 (en) 2013-02-28 2016-11-02 Univ Nanyang Tech Method of manufacturing a device for supporting biological material growth and device therefrom
US10017820B2 (en) 2013-03-05 2018-07-10 Agilent Technologies, Inc. Detection of genomic rearrangements by sequence capture
US9580746B2 (en) 2013-03-05 2017-02-28 Agilent Technologies, Inc. Synthesis of long fish probes
DK3828277T3 (en) 2013-03-13 2023-09-04 Gen9 Inc COMPOSITIONS, METHODS AND APPARATUS FOR THE SYNTHESIS OF OLIGONUCLEOTIDES
US10053719B2 (en) 2013-03-13 2018-08-21 Gen9, Inc. Compositions and methods for synthesis of high fidelity oligonucleotides
CA2906556C (en) 2013-03-15 2022-07-05 Gen9, Inc. Compositions and methods for multiplex nucleic acids synthesis
ES2946689T3 (en) 2013-03-15 2023-07-24 Univ Leland Stanford Junior Identification and use of circulating nucleic acid tumor markers
US20140274741A1 (en) 2013-03-15 2014-09-18 The Translational Genomics Research Institute Methods to capture and sequence large fragments of dna and diagnostic methods for neuromuscular disease
BR112015021521A2 (en) 2013-03-15 2017-10-10 Genentech Inc anti-crth2 antibodies and methods for their use
US20140274729A1 (en) 2013-03-15 2014-09-18 Nugen Technologies, Inc. Methods, compositions and kits for generation of stranded rna or dna libraries
US9279149B2 (en) 2013-04-02 2016-03-08 Molecular Assemblies, Inc. Methods and apparatus for synthesizing nucleic acids
US10683536B2 (en) 2013-04-02 2020-06-16 Molecular Assemblies, Inc. Reusable initiators for synthesizing nucleic acids
US9771613B2 (en) 2013-04-02 2017-09-26 Molecular Assemblies, Inc. Methods and apparatus for synthesizing nucleic acid
US20150293102A1 (en) 2013-04-13 2015-10-15 Jung-Uk Shim Detecting low-abundant analyte in microfluidic droplets
ITRM20130278A1 (en) 2013-05-10 2014-11-11 Consiglio Nazionale Ricerche PROCESS OF MANUFACTURE OF SELF-ASSEMBLED FILMS OF BLOCKED COPOLYMERS
RU2645256C2 (en) 2013-06-26 2018-02-19 Гуандун Сянсюэ Лайф Сайенсис, Лтд. High-stable t-cell receptor and method for its obtaining and application
US20150010953A1 (en) 2013-07-03 2015-01-08 Agilent Technologies, Inc. Method for producing a population of oligonucleotides that has reduced synthesis errors
KR20150005062A (en) 2013-07-04 2015-01-14 삼성전자주식회사 Processor using mini-cores
US10421957B2 (en) 2013-07-29 2019-09-24 Agilent Technologies, Inc. DNA assembly using an RNA-programmable nickase
EP3027771B1 (en) 2013-07-30 2019-01-16 Gen9, Inc. Methods for the production of long length clonal sequence verified nucleic acid constructs
ES2943498T3 (en) 2013-08-05 2023-06-13 Twist Bioscience Corp de novo synthesized libraries
US20150044648A1 (en) 2013-08-07 2015-02-12 Nike, Inc. Activity recognition with activity reminders
CN104371019B (en) 2013-08-13 2019-09-10 鸿运华宁(杭州)生物医药有限公司 It is a kind of can with GLP-1R specifically bind antibody and its with the fused protein of GLP-1
GB201314721D0 (en) 2013-08-16 2013-10-02 Almagen Ltd A method of selectively masking one or more sites on a surface and a method of synthesising an array of molecules
WO2015031689A1 (en) 2013-08-30 2015-03-05 Personalis, Inc. Methods and systems for genomic analysis
EP3044228B1 (en) 2013-09-14 2021-04-07 Chemgenes Corporation Highly efficient synthesis of long rna using reverse direction approach
WO2015040075A1 (en) 2013-09-18 2015-03-26 Genome Research Limited Genomic screening methods using rna-guided endonucleases
US9422325B2 (en) 2013-10-04 2016-08-23 Trustees Of Tufts College Glycosylation reactions using phenyl(trifluoroethyl)iodonium salts
US9582877B2 (en) 2013-10-07 2017-02-28 Cellular Research, Inc. Methods and systems for digitally counting features on arrays
US10733866B2 (en) 2013-10-29 2020-08-04 The Sheba Fund For Health Services And Research Walker-assist device
EP3063301A4 (en) 2013-10-29 2017-07-19 Longhorn Vaccines and Diagnostics, LLC Next generation genomic sequencing methods
WO2015081142A1 (en) 2013-11-26 2015-06-04 Xenco Medical, Llc Lock and release implant delivery system
WO2015081114A2 (en) 2013-11-27 2015-06-04 Gen9, Inc. Libraries of nucleic acids and methods for making the same
SG11201604495PA (en) 2013-12-04 2016-07-28 Chugai Pharmaceutical Co Ltd Antigen-binding molecules, the antigen-binding activity of which varies according to the concentration of compounds, and libraries of said molecules
CA2932325A1 (en) 2013-12-04 2015-06-11 Innovative Targeting Solutions Inc. G-protein coupled receptor agonists and methods
WO2015089053A1 (en) 2013-12-09 2015-06-18 Integrated Dna Technologies, Inc. Long nucleic acid sequences containing variable regions
EP3083692B1 (en) 2013-12-17 2020-02-19 F.Hoffmann-La Roche Ag Methods of treating her2-positive cancers using pd-1 axis binding antagonists and anti-her2 antibodies
GB2521387B (en) 2013-12-18 2020-05-27 Ge Healthcare Uk Ltd Oligonucleotide data storage on solid supports
WO2015103225A1 (en) 2013-12-31 2015-07-09 Illumina, Inc. Addressable flow cell using patterned electrodes
US9587268B2 (en) 2014-01-29 2017-03-07 Agilent Technologies Inc. Fast hybridization for next generation sequencing target enrichment
US10287627B2 (en) 2014-02-08 2019-05-14 The Regents Of The University Of Colorado, A Body Corporate Multiplexed linking PCR
WO2015134552A1 (en) 2014-03-03 2015-09-11 Swift Biosciences, Inc. Enhanced adaptor ligation
EP3116901B1 (en) 2014-03-14 2019-06-12 Immunocore Limited Tcr libraries
WO2015195178A2 (en) 2014-03-27 2015-12-23 Canon U.S. Life Sciences, Inc. Integration of ex situ fabricated porous polymer monoliths into fluidic chips
US10190161B2 (en) 2014-04-03 2019-01-29 Stmicroelectronics S.R.L. Apparatus and method for nucleic acid sequencing based on nanowire detectors
CN106232906A (en) 2014-04-15 2016-12-14 沃尔沃建造设备有限公司 Device and control method thereof for the electromotor and hydraulic pump that control engineering machinery
GB201407852D0 (en) 2014-05-02 2014-06-18 Iontas Ltd Preparation of libraries od protein variants expressed in eukaryotic cells and use for selecting binding molecules
AU2015259024B2 (en) 2014-05-16 2021-07-01 Illumina, Inc. Nucleic acid synthesis techniques
US20150361423A1 (en) 2014-06-16 2015-12-17 Agilent Technologies, Inc. High throughput gene assembly in droplets
US20150361422A1 (en) 2014-06-16 2015-12-17 Agilent Technologies, Inc. High throughput gene assembly in droplets
US10472620B2 (en) 2014-07-01 2019-11-12 General Electric Company Method, substrate and device for separating nucleic acids
US10870845B2 (en) 2014-07-01 2020-12-22 Global Life Sciences Solutions Operations UK Ltd Methods for capturing nucleic acids
US20170198268A1 (en) 2014-07-09 2017-07-13 Gen9, Inc. Compositions and Methods for Site-Directed DNA Nicking and Cleaving
WO2016011080A2 (en) 2014-07-14 2016-01-21 The Regents Of The University Of California Crispr/cas transcriptional modulation
EP3169781B1 (en) 2014-07-15 2020-04-08 Life Technologies Corporation Compositions and methods for nucleic acid assembly
WO2016022557A1 (en) 2014-08-05 2016-02-11 Twist Bioscience Corporation Cell free cloning of nucleic acids
CA2960821A1 (en) 2014-09-09 2016-03-17 Igenomx International Genomics Corporation Methods and compositions for rapid nucleic acid library preparation
WO2016053881A1 (en) 2014-10-03 2016-04-07 Life Technologies Corporation Genetic sequence verification compositions, methods and kits
CN113930455A (en) 2014-10-09 2022-01-14 生命技术公司 CRISPR oligonucleotides and gene clips
CN107109485B (en) 2014-10-10 2020-12-08 因维蒂公司 Universal blocking oligomer systems for multiple capture reactions and improved methods of hybrid capture
SG11201703138RA (en) 2014-10-18 2017-05-30 Girik Malik A biomolecule based data storage system
US10434507B2 (en) 2014-10-22 2019-10-08 The Regents Of The University Of California High definition microdroplet printer
US9890417B2 (en) 2014-11-03 2018-02-13 Agilent Technologies, Inc. Signal amplification of fluorescence in situ hybridization
US10233490B2 (en) 2014-11-21 2019-03-19 Metabiotech Corporation Methods for assembling and reading nucleic acid sequences from mixed populations
CN104562213A (en) 2014-12-26 2015-04-29 北京诺禾致源生物信息科技有限公司 Amplification sublibrary and construction method thereof
CA2975855A1 (en) 2015-02-04 2016-08-11 Twist Bioscience Corporation Compositions and methods for synthetic gene assembly
WO2016126882A1 (en) 2015-02-04 2016-08-11 Twist Bioscience Corporation Methods and devices for de novo oligonucleic acid assembly
US9834774B2 (en) 2015-02-11 2017-12-05 Agilent Technologies, Inc. Methods and compositions for rapid seamless DNA assembly
US10253363B2 (en) 2015-02-13 2019-04-09 Vaccine Research Institute Of San Diego Materials and methods to analyze RNA isoforms in transcriptomes
CN104734848A (en) 2015-03-02 2015-06-24 郑州轻工业学院 Recombinant DNA technology based information encrypting and hiding method and application
EA201792084A8 (en) 2015-04-01 2019-06-28 Зе Скриппс Рисёрч Инститьют METHODS AND COMPOSITIONS RELATED TO POLYPEPTIDES - GPCR AGONISTS
EP3280723B1 (en) 2015-04-08 2021-01-06 Polyphor AG Backbone-cyclized peptidomimetics
US11164661B2 (en) 2015-04-10 2021-11-02 University Of Washington Integrated system for nucleic acid-based storage and retrieval of digital data using keys
WO2016168755A1 (en) 2015-04-17 2016-10-20 Distributed Bio, Inc. Method for mass humanization of non-human antibodies
US9981239B2 (en) 2015-04-21 2018-05-29 Twist Bioscience Corporation Devices and methods for oligonucleic acid library synthesis
WO2016173719A1 (en) 2015-04-30 2016-11-03 Abcheck S.R.O. Method for mass humanization of rabbit antibodies
WO2016183100A1 (en) 2015-05-11 2016-11-17 Twist Bioscience Corporation Compositions and methods for nucleic acid amplification
JP6920275B2 (en) 2015-07-13 2021-08-18 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 Methods for Retrievable Information Memory Using Nucleic Acids
GB201513113D0 (en) 2015-07-24 2015-09-09 Genome Res Ltd Nasal sampling methods
GB2557529A (en) 2015-09-18 2018-06-20 Twist Bioscience Corp Oligonucleic acid variant libraries and synthesis thereof
WO2017053450A1 (en) 2015-09-22 2017-03-30 Twist Bioscience Corporation Flexible substrates for nucleic acid synthesis
US20180320166A1 (en) 2015-10-01 2018-11-08 University Of Washington Multiplex pairwise assembly of dna oligonucleotides
US20170141793A1 (en) 2015-11-13 2017-05-18 Microsoft Technology Licensing, Llc Error correction for nucleotide data stores
WO2017095958A1 (en) 2015-12-01 2017-06-08 Twist Bioscience Corporation Functionalized surfaces and preparation thereof
CN109072294A (en) 2015-12-08 2018-12-21 特温斯特兰德生物科学有限公司 For the improvement adapter of dual sequencing, method and composition
SG11201805493YA (en) 2016-01-08 2018-07-30 Iontas Ltd Binding members with altered diversity scaffold domains
GB201604492D0 (en) 2016-03-16 2016-04-27 Immatics Biotechnologies Gmbh Transfected t-cells and t-cell receptors for use in immunotherapy against cancers
KR102476915B1 (en) 2016-06-10 2022-12-12 트위스트 바이오사이언스 코포레이션 Systems and methods for automated annotation and screening of biological sequences
WO2017214557A1 (en) 2016-06-10 2017-12-14 Counsyl, Inc. Nucleic acid sequencing adapters and uses thereof
WO2018026920A1 (en) 2016-08-03 2018-02-08 Twist Bioscience Corporation Textured surfaces for polynucleotide synthesis
CN109996876A (en) 2016-08-22 2019-07-09 特韦斯特生物科学公司 The nucleic acid library of de novo formation
JP7160482B2 (en) 2016-09-02 2022-10-25 レンティジェン・テクノロジー・インコーポレイテッド Compositions and methods for treating cancer with DUOCAR
WO2018057526A2 (en) 2016-09-21 2018-03-29 Twist Bioscience Corporation Nucleic acid based data storage
JP2020504709A (en) 2016-11-18 2020-02-13 ツイスト バイオサイエンス コーポレーション Polynucleotide libraries with controlled stoichiometry and their synthesis
AU2017378492B2 (en) 2016-12-16 2022-06-16 Twist Bioscience Corporation Variant libraries of the immunological synapse and synthesis thereof
US11267883B2 (en) 2016-12-21 2022-03-08 Cephalon, Inc. Antibodies that specifically bind to human IL-15 and uses thereof
US11550939B2 (en) 2017-02-22 2023-01-10 Twist Bioscience Corporation Nucleic acid based data storage using enzymatic bioencryption
CA3056388A1 (en) 2017-03-15 2018-09-20 Twist Bioscience Corporation Variant libraries of the immunological synapse and synthesis thereof
WO2018170164A1 (en) 2017-03-15 2018-09-20 Twist Bioscience Corporation De novo synthesized combinatorial nucleic acid libraries
KR102228055B1 (en) 2017-03-23 2021-03-16 큐바이오틱스 피티와이 리미티드 Combination therapy for the treatment or prevention of tumors
US11274344B2 (en) 2017-03-30 2022-03-15 Grail, Inc. Enhanced ligation in sequencing library preparation
DK3872187T3 (en) 2017-04-23 2022-12-05 Illumina Cambridge Ltd COMPOSITIONS AND METHODS FOR IMPROVING SAMPLE IDENTIFICATION IN INDEXED NUCLEIC ACID LIBRARIES
SG11201910070PA (en) 2017-05-08 2019-11-28 Illumina Inc Universal short adapters for indexing of polynucleotide samples
WO2018231864A1 (en) 2017-06-12 2018-12-20 Twist Bioscience Corporation Methods for seamless nucleic acid assembly
WO2018231872A1 (en) 2017-06-12 2018-12-20 Twist Bioscience Corporation Methods for seamless nucleic acid assembly
CN110997116B (en) 2017-07-18 2022-08-26 百奥福瑞斯特森林研究有限公司 Method and apparatus for asymmetric polarity inversion in electromembrane processes
SG11202002194UA (en) 2017-09-11 2020-04-29 Twist Bioscience Corp Gpcr binding proteins and synthesis thereof
KR20240024357A (en) 2017-10-20 2024-02-23 트위스트 바이오사이언스 코포레이션 Heated nanowells for polynucleotide synthesis
WO2019084500A1 (en) 2017-10-27 2019-05-02 Twist Bioscience Corporation Systems and methods for polynucleotide scoring
US11427867B2 (en) 2017-11-29 2022-08-30 Xgenomes Corp. Sequencing by emergence
KR20200106067A (en) 2018-01-04 2020-09-10 트위스트 바이오사이언스 코포레이션 DNA-based digital information storage
US10722916B2 (en) 2018-01-19 2020-07-28 Caulk Garbage Can LLC Caulk gun attachment for wiping excess caulk
EP3814497A4 (en) 2018-05-18 2022-03-02 Twist Bioscience Corporation Polynucleotides, reagents, and methods for nucleic acid hybridization
US20210147830A1 (en) 2018-06-29 2021-05-20 Thermo Fisher Scientific Geneart Gmbh High throughput assembly of nucleic acid molecules
US10963953B2 (en) 2018-10-10 2021-03-30 Alliance Inspection Management, LLC Reserve management for continuous bidding portal
US10969965B2 (en) 2018-12-24 2021-04-06 Western Digital Technologies, Inc. Dynamic performance density tuning for data storage device
EP3902816A4 (en) 2018-12-26 2022-09-14 Twist Bioscience Corporation Highly accurate de novo polynucleotide synthesis
JP2022521766A (en) 2019-02-25 2022-04-12 ツイスト バイオサイエンス コーポレーション Compositions and Methods for Next Generation Sequencing
CA3131689A1 (en) 2019-02-26 2020-09-03 Twist Bioscience Corporation Variant nucleic acid libraries for glp1 receptor
CN113785057A (en) 2019-02-26 2021-12-10 特韦斯特生物科学公司 Variant nucleic acid libraries for antibody optimization
AU2020298294A1 (en) 2019-06-21 2022-02-17 Twist Bioscience Corporation Barcode-based nucleic acid sequence assembly
US20220243195A1 (en) 2019-06-21 2022-08-04 Twist Bioscience Corporation Barcode-based nucleic acid sequence assembly
EP4004232A4 (en) 2019-07-22 2023-08-09 Igenomx International Genomics Corporation Methods and compositions for high throughput sample preparation using double unique dual indexing
WO2021046655A1 (en) 2019-09-13 2021-03-18 University Health Network Detection of circulating tumor dna using double stranded hybrid capture
EP4034566A4 (en) 2019-09-23 2024-01-24 Twist Bioscience Corp Variant nucleic acid libraries for crth2
AU2020355027A1 (en) 2019-09-23 2022-04-21 Twist Bioscience Corporation Variant nucleic acid libraries for single domain antibodies
AU2020400030A1 (en) 2019-12-09 2022-07-07 Twist Bioscience Corporation Variant nucleic acid libraries for adenosine receptors
US20210395344A1 (en) 2020-04-27 2021-12-23 Twist Bioscience Corporation Variant nucleic acid libraries for coronavirus
EP4179001A2 (en) 2020-07-07 2023-05-17 Twist Bioscience Corporation Devices and methods for light-directed polymer synthesis
WO2022046797A1 (en) 2020-08-25 2022-03-03 Twist Bioscience Corporation Compositions and methods for library sequencing
JP2023539245A (en) 2020-08-26 2023-09-13 ツイスト バイオサイエンス コーポレーション Methods and compositions related to GLP1R variants
US20220064206A1 (en) 2020-08-28 2022-03-03 Twist Bioscience Corporation Devices and methods for synthesis
AU2021358892A1 (en) 2020-10-05 2023-06-08 Twist Bioscience Corporation Hybridization methods and reagents
EP4229210A1 (en) 2020-10-19 2023-08-23 Twist Bioscience Corporation Methods of synthesizing oligonucleotides using tethered nucleotides
WO2022087293A1 (en) 2020-10-22 2022-04-28 Twist Bioscience Corporation Methods and systems for detecting coronavirus
WO2022093811A1 (en) 2020-10-26 2022-05-05 Twist Bioscience Corporation Libraries for next generation sequencing
WO2022098662A2 (en) 2020-11-03 2022-05-12 Twist Bioscience Corporation Methods and compositions relating to chemokine receptor variants
CA3206018A1 (en) 2021-01-21 2022-07-28 Twist Bioscience Corporation Methods and compositions relating to adenosine receptors
US20220277808A1 (en) 2021-02-19 2022-09-01 Twist Bioscience Corporation Libraries for identification of genomic variants
WO2022204301A1 (en) 2021-03-24 2022-09-29 Twist Bioscience Corporation Electrochemical polynucleotide synthesis
EP4314075A2 (en) 2021-03-24 2024-02-07 Twist Bioscience Corporation Variant nucleic acid libraries for cd3
EP4314074A1 (en) 2021-03-24 2024-02-07 Twist Bioscience Corporation Variant nucleic acid libraries for tigit
CA3214947A1 (en) 2021-04-09 2022-10-13 Twist Bioscience Corporation Libraries for mutational analysis
US20220411784A1 (en) 2021-05-03 2022-12-29 Twist Bioscience Corporation Variant nucleic acid libraries for glycans
EP4334516A1 (en) 2021-05-03 2024-03-13 Twist Bioscience Corporation Variant nucleic acid libraries for ion channels

Also Published As

Publication number Publication date
US20160310927A1 (en) 2016-10-27
US20180236425A1 (en) 2018-08-23
US11691118B2 (en) 2023-07-04
US20200330953A1 (en) 2020-10-22
US9981239B2 (en) 2018-05-29
WO2016172377A1 (en) 2016-10-27
US10744477B2 (en) 2020-08-18

Similar Documents

Publication Publication Date Title
US11691118B2 (en) Devices and methods for oligonucleic acid library synthesis
US11697668B2 (en) Methods and devices for de novo oligonucleic acid assembly
US20180104664A1 (en) Textured surfaces for polynucleotide synthesis
US20240043920A1 (en) Polynucleotides, reagents, and methods for nucleic acid hybridization
US11562103B2 (en) Nucleic acid based data storage
US10987648B2 (en) Functionalized surfaces and preparation thereof
US20230193383A1 (en) Flexible substrates for nucleic acid synthesis
US20160251651A1 (en) Cell free cloning of nucleic acids
US11377676B2 (en) Methods for seamless nucleic acid assembly

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: APPLICATION UNDERGOING PREEXAM PROCESSING

STCB Information on status: application discontinuation

Free format text: ABANDONED -- INCOMPLETE APPLICATION (PRE-EXAMINATION)