NZ795976A - Binding polypeptides and methods of making the same - Google Patents

Binding polypeptides and methods of making the same

Info

Publication number
NZ795976A
NZ795976A NZ795976A NZ79597617A NZ795976A NZ 795976 A NZ795976 A NZ 795976A NZ 795976 A NZ795976 A NZ 795976A NZ 79597617 A NZ79597617 A NZ 79597617A NZ 795976 A NZ795976 A NZ 795976A
Authority
NZ
New Zealand
Prior art keywords
cell
strand
sequence
cdna
chain
Prior art date
Application number
NZ795976A
Inventor
Gregory Babcock
Luke Robinson
Zachary Shriver
Original Assignee
Visterra Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Visterra Inc filed Critical Visterra Inc
Publication of NZ795976A publication Critical patent/NZ795976A/en

Links

Abstract

Polypeptides, such as antibody molecules and TCR molecules, and methods of making the same, are disclosed. The polypeptides can be used to treat, prevent, and/or diagnose disorders.

Description

Polypeptides, such as antibody les and TCR molecules, and methods of making the same, are disclosed. The polypeptides can be used to treat, prevent, and/or diagnose disorders.
NZ 795976 BINDING POLYPEPTIDES AND METHODS OF MAKING THE SAME This is a onal ofNew Zealand patent application No. 753978, the entire contents of which are incorporated herein by reference.
BACKGROUND Monoclonal antibody therapies are a class of irnmunotherapies that involve monoclonal antibodies (mAbs) that are capable of specifically cting with e-relevant biological molecules. In recent years, the disease areas that therapeutic antibodies can target have significantly expanded, and a number of monoclonal antibodies and antibody-derivative products have been ed for therapeutic use in the United States and many other countries. Monoclonal antibody therapies are currently used or investigated for treating various diseases or conditions, including, for example, infectious diseases, cancer, immune diseases, organ transplantation, cardiovascular diseases, and metabolic diseases.
Given the y of monoclonal antibodies and dy-derivative ts in modulating various biological functions, the need exists for developing new approaches for generation of antibodies le for treating, preventing, and diagnosing disorders.
SUMMARY This disclosure provides, at least in part, binding polypeptides (e.g., antibody molecules or T- cell receptor (TCR) molecules) that comprise one or more of the structural or functional properties disclosed herein. In an embodiment, libraries of the binding polypeptides, methods for making the polypeptides or libraries, nucleic acid molecules encoding the binding polypeptides, expression vectors, host cells, compositions (e.g., ceutical compositions), kits, and containers, are also ed. The ptides (e.g., antibody molecules or TCR molecules) disclosed herein can be used (alone or in combination with other agents or therapeutic modalities) to treat, prevent and/or diagnose disorders, such as disorders and conditions disclosed herein.
In an aspect, the disclosure features a method of making a nucleic acid sequence comprising a sequence that encodes a heavy chain element (HC t) of an antibody heavy chain variable region (HCVR) and a light chain t (LC element) of an antibody light chain variable region (LCVR), and wherein the HCVR and LCVR are matched, the method comprising: a) acquiring an ed tion reaction site, e.g., a production micro-chamber, comprising: i) a heavy chain (HC) strand, wherein the HC strand is a strand of a heavy chain - ed cDNA (HC ds cDNA) comprising a segment that encodes an HC element of the HCVR from a cell, e. g., a heavy chain le region sequence (HCVRS); and ii) a light chain (LC) strand, n the LC strand is a strand of a light chain double-stranded cDNA (LC ds cDNA) comprising a segment that encodes an LC element of the LCVR from the cell, e.g., a light chain variable region sequence (LCVRS), and b) covalent linking, e. g., ligation, of an HC strand to an LC strand, wherein the isolated production reaction site, e.g., a tion micro-chamber, does not e a nucleic acid encoding an HCVR or an LCVR from a cell other than the cell (e. g., a different cell, e.g., a different B cell), thereby making a nucleic acid sequence comprising a sequence that encodes an HC element of an HCVR and a LC element of an LCVR, wherein the HCVR and LCVR are matched.
In an embodiment, the HC element comprises, or consists of, an HCVRS, or a onal nt thereof (e.g., an antigen binding fragment thereof). In an embodiment, the LC element comprises, or ts of, an LCVRS, or a functional fragment thereof (e.g., an antigen binding fragment thereof).
In an embodiment, the HC ds cDNA comprises a segment that encodes an HCVRS. In an embodiment, the LC ds cDNA comprises a segment that encodes an LCVRS. In an embodiment, the HC ds cDNA comprises a t that s an HCVRS, and the LC ds cDNA comprises a segment that encodes an LCVRS.
In an embodiment, the cell is an immune cell, e.g., a B cell, e.g., a human B cell. In an embodiment, the cell is a mammalian cell or an avian cell.
In an ment, the nucleic acid sequence is configured such that, when expressed, the HC element and the LC element (6.3., the HCVRS and the LCVRS) form a functional antigen binding molecule, e.g., an scFv, an Fab, or an scFab. In an embodiment, the antigen binding molecule, e.g., an scFv, is functional in vitro, ex vivo, or in vivo, e.g., as determined by a method or assay described herein.
In an embodiment, acquiring an isolated production reaction site, e.g., a production micro- chamber, comprises: a) ing a capture substrate bound to: (i) a first double-stranded cDNA (ds cDNA) comprising a strand that is mentary to a first mRNA that encodes an HCVR from a cell; and (ii) a second ds cDNA comprising a strand complementary to a second mRNA encoding an LCVR from the cell (the cDNA loaded capture substrate), and b) maintaining the isolated production reaction site, e.g., the production micro-chamber, under conditions that allow amplification of the first and second ds cDNAs, to produce: a plurality of HC ds cDNAs comprising a segment that encodes an HC element of the HCVR from the cell, e.g., an HCVRS; and a plurality of LC ds cDNAs comprising a segment that encodes an LC element of the LCVR from the cell, e. g., an LCVRS.
In an embodiment, the HC ds cDNA is identical, or substantially identical, to the first ds cDNA. For example, the sense strand of the HC ds cDNA is at least 80%, 85 %, 90%, 95%, 98%, 99%, or 100% cal to, or differs by no more than 1, 2, 5, 10, 15, 20, 25, 30, 35, 40, 45, or 50 nucleotides from, the sense strand of the first ds cDNA, and/or the antisense strand of the HC ds cDNA is at least 80%, 85%, 90%, 95%, 98%, 99%, or 100% identical to, or differs by no more than 1, 2, 5, 10, 15, 20, 25, 30, 35, 40, 45, or 50 nucleotides from, the antisense strand of the first ds cDNA.
In an embodiment, the LC ds cDNA is identical, or substantially identical, to the second ds cDNA. For example, the sense strand of the LC ds cDNA is at least 80%, 85%, 90%, 95%, 98%, 99%, or 100% identical to, or differs by no more than 1, 2, 5, 10, 15, 20, 25, 30, 35, 40, 45, or 50 nucleotides from, the sense strand of the second ds cDNA, and/or the antisense strand of the LC ds cDNA is at least 80%, 85%, 90%, 95%, 98%, 99%, or 100% identical to, or differs by no more than 1, 2, 5, 10, 15, 20, 25, 30, 35, 40, 45 , or 50 tides from, the antisense strand of the second ds cDNA.
In an embodiment, the HC strand is a sense strand. In an embodiment, the LC strand is a sense strand. In an embodiment, the HC strand is an antisense strand. In an embodiment, the LC strand is an antisense strand. In an embodiment, both the HC strand and the LC strand are sense strands. In an embodiment, both the HC strand and the LC strand are antisense strands.
In an embodiment, the capture substrate comprises a bead, e.g., a magnetic bead. In an embodiment, the capture substrate comprises a moiety (e.g., an oligonucleotide) which binds to cDNA, e.g., (i) a moiety which binds to the HC strand; (ii) a moiety which binds to the LC ; or (iii) both (i) and (ii). In an embodiment, the moiety which binds to the HC strand is different from the moiety which binds to the LC strand, e.g., to facilitate creating conditions favorable to ing similar levels of each DNA le type. In an embodiment, the moiety which binds to the HC strand is identical to the moiety which binds to the LC strand.
In an embodiment, the first mRNA and the second mRNA are disposed on an mRNA loaded capture substrate.
In an embodiment, the isolated production reaction site, e.g., the production micro-chamber, comprises: a reagent mixture suitable for producing, from the first and second mRNAs (e.g., after the first and second mRNAs are released from the mRNA loaded capture substrate into a solution), a first ds cDNA comprising a segment that encodes an HC element of the HCVR of the cell, e.g., an HCVRS, and a second ds cDNA comprising a segment that s an LC element of the LCVR of the cell, e.g., an LCVRS.
In an embodiment, the isolated production on site, e.g., tion micro-chamber, comprises primers that mediate the production of the first ds cDNA. In an embodiment, the isolated production on site, e.g., production micro-chamber, comprises primers that mediate the production of the second ds cDNA.
In an embodiment, a cDNA strand that is complementary to a first mRNA that encodes an HCVR from a cell is made by reverse transcription of the first mRNA. In an embodiment, a cDNA strand that is mentary to a second mRNA that encodes an LCVR from a cell is made by reverse transcription of the second mRNA.
In an embodiment, the reverse transcription takes place in the isolated production reaction site, e.g., a production-micro chamber. In an ment, the reverse transcription takes place in an isolated cell reaction site, e.g., a cell isolation micro-chamber. In an ment, the reverse transcription takes place outside the isolated production reaction site, e.g., a production micro- chamber, or outside an isolated cell reaction site, e.g., a cell isolation micro-chamber. In an embodiment, the reverse transcription takes place outside the ed production reaction site, e.g., a production-micro chamber, and outside an isolated cell reaction site, e.g., a cell isolation micro- chamber. In an embodiment, the reverse transcription takes place outside an isolated reaction site, e.g., outside a chamber.
In an embodiment, the amplification comprises 30 or fewer cycles, e.g., 20 or fewer cycles, e.g., 15 or fewer, 14 or fewer, 13 or fewer, 12 or fewer, 11 or fewer, 10 or fewer, 9 or fewer, 8 or fewer, 7 or fewer, 6 or fewer, or 5 or fewer cycles.
In an embodiment, the reverse transcription and/or amplification uses one or more primers, e.g., comprising a sequence specific for an HCVRS and/or an LCVRS.
In an embodiment, the reverse transcription and/or amplification comprises using two or more primers that mediate the production of the HC ds cDNA, wherein at least one primer comprises a nucleotide modification, and wherein at least one primer does not comprise a nucleotide modification.
In an embodiment, the amplification ses using two or more primers that mediate the tion of the LC ds cDNA, wherein at least one primer ses a nucleotide modification, and wherein at least one primer does not comprise a nucleotide modification.
In an embodiment, at least one primer comprises a tide modification, e.g., which reduces, e.g., inhibits, DNA synthesis, e.g., by a DNA polymerase. In an embodiment, at least one primer does not comprise a nucleotide modification, e.g., which reduces, e.g., inhibits, DNA synthesis, e.g., by a DNA polymerase.
In an embodiment, the nucleotide modification inhibits a DNA rase from extending the DNA. Without wishing to be bound by theory, it is believed that in an embodiment, any chemical entity that reduces (e.3., blocks) DNA polymerase extension can be used in accordance with the methods described herein.
In an ment, the nucleotide modification is an insertion of a spacer to the , e. g., between two nt nucleotides in the primer. In an embodiment, the spacer is a flexible spacer. In an embodiment, the spacer is a carbon spacer (e.g., n-, wherein n=3, 4, 5, 6, 7, 8, 9, 10, or more), two or more (e.g., three, four, five, six, seven, eight, nine, ten, or more) abasic nucleotides, or a hylene glycol (PEG) spacer. In an embodiment, the spacer is a PEG . In an embodiment, the tide modification is 2’-O-methyl, 2’-OH, 2’-NH2, or uracil, e.g., to a ribose.
In an embodiment, the nucleotide modification is located internally or at the 3’ end of the . In an embodiment, at least one primer comprises (i) a first member; (ii) a second member; and optionally (iii) a third member, e.g., comprising a nucleotide modification described herein, e.g., d between (i) and (ii).
In an embodiment, the first member is capable of annealing with the second member. In an embodiment, the first member is capable of annealing with the second member in the same primer, e.g., through intra-molecular hybridization, e.g., to form a hairpin structure sing a duplex region of 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, more basepairs. In another embodiment, the first member is capable of annealing hybridizing with the second member in a different primer, e.g., through inter-molecular hybridization, e.g., to form a double-stranded structure sing a duplex region of 4, 5,6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, more basepairs. Without wishing to be bound by theory, it is believed that in an ment, there are at least two secondary structures that the modified primers can form and facilitate reduction (e.g., prevention) of competition to substrate (e.g., bead) capture. For example, the secondary structure can be a hairpin-like structure formed by intra-molecular hybridization (within the same primer), or the secondary structure can be a duplex structure formed by molecular hybridization (between two different s).
In an embodiment, the first member comprises a sequence that is mentary to the sequence of an ucleotide attached to the capture substrate. In an embodiment, the second member comprises (e.g., from 5’ to 3’) one, two, or all of: (i) a sequence that is mentary to at least a portion of the first member; (ii) a universal priming sequence (e.g., for PCR amplification or next-generation sequencing); and (iii) a sequence complementary to a target sequence, e.g., an HCVRS and/or an LCVRS. In an embodiment, the universal priming sequence is identical, or substantially identical, to the sequence that is complementary to at least a portion of the first member.
In another embodiment, the universal priming ce is ent from the sequence that is complementary to at least a portion of the first member. In an embodiment, the second member comprises a sequence for homologous recombination (e.g., in a yeast or mammalian cell).
In an embodiment, at least one primer comprises a sequence encoding at least a portion of a linker sequence, or a complementary sequence thereof. In an ment, the primer that ses a sequence encoding at least a portion of a linker sequence, or a complementary sequence f, is phosphorylated, e.g., 5’ phosphorylated. Without wishing to be bound by theory, it is believed that in an embodiment, any sequence with the general properties of flexibility (e.g., facilitated by glycine) and hydrophilicity can work effectively in accordance with the methods described herein. Exemplary linkers can generally have overrepresentation of one or more of Gly, Ser, Thr, or Ala and WO 19402 underrepresentation of hydrophobic residues, e.g., one or more of Trp, Tyr, Phe, Cys, Met, Leu, or Ile.
The length of the primer may vary, e.g., 3-50 amino acid es (e.g., 5-45, 10-40, 15-35, 20-30, 10- , 10-30, 20-40, or 30-40 amino acid residues). In an embodiment, the linker sequence comprises, or consists of, ((Gly)m-Ser))n, where m=3, 4, 5, or more and n=1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more. In an embodiment, the linker sequence comprises, or consists of, (Gly-Gly-Gly-Gly-Ser)n, where n=1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more.
In an embodiment, the primer is a primer described herein, e.g., in Examples.
In an embodiment, the reverse transcription, the amplification, or both, occurs in a solution in the isolated production on site, e.g., production micro-chamber. In an embodiment, the reverse transcription, the amplification, or both, does not occur on the substrate (e.g., bead). For example, the reverse transcription, the amplification, or both, can occur on in a solution within a droplet.
In an embodiment, the HC ds cDNA comprises a 5’ overhang, e.g., a 5’ overhang that is capable of hybridizing to an oligonucleotide attached to a capture substrate. In an embodiment, the HC ds cDNA comprises a blunt end, e.g., a blunt end comprising a 5’ phosphate. In an embodiment, the LC ds cDNA comprises a 5’ overhang, e.g., a 5’ overhang that is capable of izing to an oligonucleotide attached to a capture substrate. In an embodiment, the LC ds cDNA comprises a blunt end, e.g., a blunt end comprising a 5’ phosphate. In an ment, the HC ds cDNA and the LC ds cDNA comprise sticky ends, e.g., both have 5 ’ overhangs.
In an embodiment, the HC strand and the LC strand are covalently linked, e.g., ligated, to produce a single stranded nucleic acid sequence, wherein the HC and LC s are both sense s or both antisense s. In an ment, a denatured HC strand of the HC ds cDNA to a denatured LC strand of the LC ds cDNA are covalently , e.g., ligated, wherein the HC and LC strands are both sense strands or both antisense strands. In an embodiment, the HC strand is present in the HC ds cDNA and the LC strand is present in the LC ds cDNA, and wherein the HC ds cDNA and the LC ds cDNA are covalently linked, e.g., ligated, e.g., to produce a double stranded nucleic acid sequence.
In an embodiment, the covalent linking, e. g., on, occurs in the ed production reaction site. In an embodiment, the isolated production reaction site, e.g., a production micro- chamber, or the isolated linkage reaction site, e.g., a linkage micro-chamber, comprises a reagent that is e of covalently linking, e.g., ligating, the HC and LC strands or the HC and LC ds cDNAs.
In an embodiment, the isolated production reaction site, e.g., a production micro-chamber ses an enzyme that covalently couples the HC and LC strands or the HC and LC ds cDNAs. In an embodiment, the enzyme is a ligase, e. g., a thermal stable ligase. In an embodiment, the covalent linking comprises ligase thermocycling.
In an embodiment, the covalent linking, e. g., ligation, occurs in a site different from the isolated production reaction site, e.g., occurs in an isolated e reaction site, e.g., a linkage micro- chamber. In an embodiment, the HC strand and the LC strand are transferred from the isolated production site to the ed linkage reaction site, e.g., a linkage micro-chamber, and the covalent linking occurs in the isolated linkage reaction site, e.g., a linkage micro-chamber. In an embodiment, the isolated linkage on site, e.g., a linkage micro-chamber, comprises a reagent that is capable of covalently linking, e.g., ligating, the HC and LC strands or the HC and LC ds cDNAs. In an embodiment, the ed linkage on site, e. g., a linkage micro-chamber, comprises an enzyme that covalently s the HC and LC s or the HC and LC ds cDNAs. In an embodiment, the enzyme is a , e.g., a thermal stable ligase. In an embodiment, the covalent linking comprises ligase thermocycling.
In an embodiment, the covalent linking, e.g., ligation, comprises: (a) heating the isolated linkage reaction site, e.g., the linkage micro-chamber, under conditions (e.g., at 950C) that allow denaturation of the HC strand and the LC strand; (b) cooling the isolated linkage reaction site, e.g., the linkage micro-chamber, under conditions (e.g., at 50-65°C) that allow hybridization of the splint oligonucleotide to the HC strand and the LC strand; (c) maintaining the isolated linkage reaction site, e. g., the linkage micro-chamber, under ions (e.g., at 45-65°C) that allow ligation of the HC strand and the LC strand (e.g., formation of phosphodiester bond between the HC strand and the LC strand); and (d) repeating steps (a), (b), and (c) sequentially for 2, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, or more cycles.
In an embodiment, the HC strand and the LC strand are covalently linked, e.g., ligated, in the presence of a splint oligonucleotide. In an embodiment, the splint oligonucleotide is hybridized to a sequence comprising the junction of the HC strand and the LC strand, or a sequence complementary thereof, and forms a duplexed region at the site of on. In an ment, the splint oligonucleotide comprises a modification (e.g., an NH; group) that inhibits DNA synthesis, e.g., by a DNA polymerase. In an embodiment, the modification is at the 3’ end of the splint oligonucleotide.
In an embodiment, a strand complimentary to the ntly linked, e.g., ligated, HC and LC strands is produced by amplification.
In an embodiment, the method, e.g., the step of covalent linkage, does not include a step of overlap extension polymerase chain reaction (OE-PCR), also known as splicing by p extension or splicing by overhang extension (SOE) PCR.
In an embodiment, the method further comprises, prior to ing the isolated production reaction site, e.g., a production micro-chamber, acquiring an mRNA loaded capture ate.
In an embodiment, acquiring the mRNA loaded e substrate comprising: a) acquiring an isolated cell reaction site, e.g., a cell isolation micro-chamber, comprising: i) a cell; and ii) a capture substrate capable of binding a first mRNA encoding an HCVR from the cell and a second mRNA encoding an LCVR from the cell; and b) maintaining the isolated cell reaction site, e.g., the cell isolation micro-chamber, under conditions that allow lysis of the cell and binding of the capture substrate with the first mRNA and the second mRNA to form the mRNA loaded e substrate, wherein the isolated cell reaction site, e.g., cell isolation micro-chamber, does not include a nucleic acid encoding an HCVR or an LCVR from a cell other than the cell (e.g., a different cell).
In an ment, the isolated cell reaction site, e.g., cell isolation micro-chamber, comprises a lysing reagent, e.g., a detergent. In an embodiment, the cell is lysed by heat or an enzyme. In an ment, the capture substrate comprises a moiety (e.g., an oligonucleotide) which binds mRNA, 6.3., an dT).
In an embodiment, the method further comprises releasing the mRNA loaded capture substrate from the ed cell reaction site, e.g., the cell isolation micro-chamber. In an embodiment, the releasing step is performed in the presence of a poly(dA) or T) oligonucleotide, e.g., to reduce cross-binding of non-captured mRNA.
In an embodiment, the mRNA loaded capture substrate is transferred from the isolated cell reaction site, e.g., the cell isolation micro-chamber, to the isolated production reaction site, e.g., the production micro-chamber.
In an embodiment, the method further comprises releasing the c acid sequence from the isolated tion reaction site, e.g., the tion micro-chamber. In an embodiment, the method further comprises amplifying the nucleic acid sequence. In an ment, amplification of the nucleic acid sequence occurs outside the isolated production reaction site, e.g., the production micro- chamber, e.g., after the nucleic acid is ed from the isolated production reaction site, e.g., the production chamber. In an embodiment, amplification of the nucleic acid sequence occurs at the ed tion reaction site, e. g., the production micro-chamber.
In an embodiment, the method further comprises sequencing all or a portion of the nucleic acid sequence.
In an embodiment, the method further comprises inserting all or a portion of nucleic acid sequence into a vector. In an embodiment, the vector supplies an additional HC element or LC element not included in the nucleic acid sequence. In an embodiment, the vector supplies an HC CDRl, an HC CDRZ, or both. In an embodiment, the method further comprises sing the vector.
In an embodiment, the method r comprises expressing the nucleic acid sequence to produce a polypeptide comprising a segment that encodes an HC element of the HCVR, e.g., an HCVRS, and a segment that encodes an LC element of the LCVR, e.g., an LCVRS. In an embodiment, the LC element is inal to the HC element in the polypeptide. In an embodiment, the HC element is C-terminal to the LC element in the polypeptide.
In an embodiment, the method further comprises contacting the polypeptide with an antigen.
In an embodiment, the method further ses determining if the polypeptide binds the antigen, in vitro, ex vivo, or in viva, e.g., by a method or assay described herein.
In an aspect, the sure features a method of making a nucleic acid sequence comprising a sequence that encodes a heavy chain element (HC element) of an antibody heavy chain variable region (HCVR) and a light chain t (LC element) of an antibody light chain variable region (LCVR), and wherein the HCVR and LCVR are matched, comprising: a) acquiring an isolated cell reaction site (e.g., an isolated cell reaction site described herein), 6.3., a cell isolation micro-chamber, comprising: i) a cell (e.g., a cell described herein); and ii) a capture substrate (e. g., a e substrate described herein) capable of binding a first mRNA encoding an HCVR from the cell and a second mRNA encoding an LCVR from the cell; b) maintaining the isolated cell reaction site, e.g., the cell isolation micro-chamber, under conditions that allow lysis of the cell and binding of the e substrate with the first mRNA and the second mRNA to form an mRNA loaded capture substrate, wherein the isolated cell reaction site, e.g., cell ion micro-chamber, does not include a nucleic acid encoding an HCVR or an LCVR from a cell other than the cell (e.g., a different cell); c) contacting the mRNA loaded capture substrate with a reaction mixture, e.g., a reaction mixture comprising reverse transcriptase, that uses the loaded mRNA as a template to make cDNA (this can occur, e.g., in the isolated cell reaction site, in an isolated production reaction site, or in neither, e.g., not in an isolated reaction site); d) acquiring an isolated production reaction site (e.g., an isolated production on site described herein), e.g., a production chamber, comprising: i) a heavy chain (HC) strand, wherein the HC strand is a strand of a heavy chain double-stranded cDNA (HC ds cDNA) comprising a segment that encodes an HC element of the HCVR from the cell, e.g., a heavy chain variable region sequence (HCVRS); and ii) a light chain (LC) strand, wherein the LC strand is a strand of a light chain double-stranded cDNA (LC ds cDNA) comprising a segment that encodes an LC element of the LCVR from the cell, e.g., a light chain variable region sequence ), wherein the isolated production reaction site, e.g., a tion micro-chamber, does not include a nucleic acid encoding an LCVR or an HCVR from a cell other than the cell (e.g., a different cell); and e) nt linking, e.g., ligation, of the HC strand to the LC strand.
In an embodiment, one or more (e.g., two, three, four, or all) of the steps a)-e) are med in accordance with a method described . In an ment, each of the steps a)-e) is performed in accordance with a method described herein.
In an aspect, the disclosure features a method of making a nucleic acid ce comprising a sequence that encodes a heavy chain t (HC element) of an antibody heavy chain variable region (HCVR) and a light chain element (LC element) of an antibody light chain variable region (LCVR), and wherein the HCVR and LCVR are matched, comprising: a) acquiring an isolated cell reaction site (e.g., an isolated cell reaction site described herein), e.g., a cell isolation micro-chamber, comprising: i) a cell (e.g., a cell described herein); and ii) a capture ate (e. g., a capture substrate described herein) capable of binding a first mRNA encoding an HCVR from the cell and a second mRNA encoding an LCVR from the cell; b) ining the isolated cell reaction site, e.g., the cell isolation micro-chamber, under conditions that allow lysis of the cell and binding of the capture substrate with the first mRNA and the second mRNA to form the mRNA loaded capture substrate, wherein the isolated cell reaction site, e.g., cell isolation micro-chamber, does not include a c acid encoding an HCVR or an LCVR from a cell other than the cell (e.g., a different cell); c) acquiring an isolated production reaction site (e.g., an isolated production reaction site described herein), e.g., a production micro-chamber, comprises: contacting the mRNA loaded capture substrate with a reaction mixture, e.g., a reaction mixture comprising reverse transcriptase, that uses the loaded mRNA as a te, to produce: a first double-stranded cDNA (ds cDNA) comprising a strand that is complementary to a first mRNA that s an HCVR from a cell; and a second ds cDNA comprising a strand complementary to a second mRNA encoding an LCVR from the cell (the cDNA loaded capture substrate); wherein the isolated production reaction site, e.g., a production micro-chamber, does not include a nucleic acid ng an LCVR or an HCVR from a cell other than the cell (e. g., a different cell). d) ining the isolated production reaction site, e.g., the production micro-chamber, under conditions that allow amplification of the first and second ds cDNAs, to produce: a ity of HC ds cDNAs comprising a t that encodes an HC element of the HCVR from the cell, e. g., an HCVRS; and a plurality of LC ds cDNAs comprising a t that encodes an LC element of the LCVR from the cell, e.g., an LCVRS; e) acquiring an isolated linkage reaction site (e.g., an isolated e reaction site described herein), e.g., a linkage micro-chamber, comprising: covalent linking, e.g., ligation, of a strand of the HC ds cDNA (HC strand) to a strand of the LC ds cDNA (LC strand), wherein the HC and LC strands are both sense strands or antisense strands; and t) amplifying the covalently , e.g., d, HC and LC s.
In an embodiment, one or more (e.g., two, three, four, five, or all) of the steps a)-f) are performed in accordance with a method described herein. In an embodiment, each of the steps a)-f) is performed in ance with a method described herein.
In an aspect, the disclosure features a method of making a library comprising a plurality of unique members, the method comprising: making the plurality of members, wherein each of the members ses a sequence that encodes a heavy chain element (HC element) of a heavy chain variable region (HCVR) and a light chain element (LC element) of a light chain variable region (LCVR), and wherein the HCVR and LCVR are matched, made by a method described herein, wherein each unique nucleic acid sequence of the plurality ses an HC t and an LC element from a ent unique cell (e.g., a cell described herein), thereby making a library comprising a plurality of unique members.
In an embodiment, the plurality of unique members ses at least 104, 105, 106, 107, 108, or 109 unique members. In an embodiment, the plurality of unique members comprises 104 to 109, 104 to 10“, 104 to 107, 104 to 106, 104 to 105, 108 to 109, 107 to 109, 106 to 109, 105 to 109, 105 to 108, 106 to 107, 104 to 105, 105 to 106, 106 to 107, 107 to 108, or 108 to 109 unique members. In an embodiment, at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%, of the members in the library are unique members (which encode matched HC element and LC element sequences). In an embodiment, less than 20%, 15%, 10%, 5%, 4%, 3%, 2%, or 1%, of the members in the library are unique members (which encode matched HC element and LC element sequences).
In an aspect, the disclosure features a y comprising a plurality of unique members, wherein, i) each unique member of the plurality comprises a segment that encodes an HC element, e.g., an HCVRS, and a segment that encodes an LC t, e. g., an LCVRS, wherein the HC element and the LC element in each unique member is matched; ii) each unique member of the ity comprises a segment that encodes an HC element, 6.3., an HCVRS, and a segment that encodes an LC t, e.g., an LCVRS, from a different unique cell; and iii) the library comprises one or more (e.g., two, three, four, or all) of the following properties: a) the library is made by a method described herein; b) the plurality of unique members comprises at least 104, 105, 106, 107, 108, or 109 unique nucleic acid sequences; c) the plurality of unique s comprises 104 to 109, 104 to 108, 104 to 107, 104 to 106, 104 to 105, 108 to 109, 107 to 109, 106 to 109, 105 to 109, 105 to 108, 106 to 107, 104 to 105, 105 to 106, 106 to 107, 107 to 108, or 108 to 109 unique members; d) at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%, of the members in the y are unique members (which encode matched HC element and LC element sequences); or e) less than 20%, 15%, 10%, 5%, 4%, 3%, 2%, or 1%, of the members in the library are unique s (which encode matched HC element and LC element sequences).
In an ment, each unique member of the plurality is configured such that, when expressed, the HC element, e.g., the HCVRS, and the LC element, e.g., the LCVRS, form a functional antigen binding molecule, e.g., an scFv, an Fab, or an scFab.
In an embodiment, the library is a display library. In an embodiment, each of the members of the plurality further encodes a polypeptide that results in display of the member on the surface of a display entity. In an embodiment, the library is a phage display library. In an embodiment, the y is a yeast display y. In an embodiment, the library is a mammalian display library.
In an aspect, the disclosure features a method of making a binding polypeptide (e.g., a polypeptide comprising an HC element and an LC element), the method comprising: a) acquiring a y described herein, e.g., by a method described herein; and b) expressing a polypeptide encoded by a unique nucleic acid of the library.
In an embodiment, the method further ses contacting the polypeptide with an antigen.
In an embodiment, the method further comprises retrieving (e.g., isolating or purifying) the nucleic acid that s a polypeptide that binds the antigen.
In an aspect, the disclosure features an isolated production reaction site, e.g., a production micro-chamber, which is an isolated production reaction site described herein (e.g., sing a nucleic acid encoding an HCVR and a nucleic acid encoding a LCVR, wherein the HCVR and the LCVR are matched).
In an embodiment, the isolated production on site, e.g., a production chamber, does not include a nucleic acid encoding an HCVR or an LCVR from a different cell.
In an embodiment, the isolated production reaction site, e.g., a production micro-chamber, comprises one, two, or all of: (i) one or more s specific to V gene sequences of the HC and LC; (ii) one or more primers specific to overhangs uced onto the HC and LC cDNAs; or (iii) one or more primers comprising a first member, a second member, and a third member comprising a nucleotide modification (e.g., a ) located between the first and second members, wherein the first member is capable of annealing with the second member of the same primer or a different , e.g., forming a structure comprising a duplex region of 4, 5, 6, 7, 8, 9, 10, ll, 12, l3, 14, 15, 16, 17, 18, 19, 20, more basepairs.
In an embodiment, the ed production reaction site, e. g., a production micro-chamber, does not comprise a reagent that can covalently link nucleic acids, e.g., a ligase, e.g., a thermostable ligase. In another embodiment, the isolated tion reaction site, e.g., a production micro- chamber, comprises a reagent that can covalently link c acids, e.g., a ligase, e.g., a thermostable 1i gase.
In an aspect, the sure features a self-annealing oligonucleotide comprising a first member, a second member, and third member comprising a nucleotide modification (e.g., a spacer) located n the first and second members, n the first member is capable of annealing with the second member of the same oligonucleotide (e.g., for a method of making a nucleic acid sequence comprising a sequence that encodes an HC element of an HCVR and a LC element of an LCVR, wherein the HCVR and LCVR are matched).
In an ment, the first and second members are e of forming a hairpin structure comprising a duplex region of 4, 5,6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, more basepairs. In an embodiment, the first member is 5-40 nucleotides, 6.3., 5-10, 5-20, 5-30, 30-40, 20— 40, 10-30, 10-30, or 15-25 nucleotides, in length. In an embodiment, the second member is 5-40 nucleotides, e.g., 5-10, 5-20, 5-30, 30-40, 20-40, 10-30, 10-30, or 15-25 nucleotides, in length.
In an embodiment, the spacer is a spacer described herein, e.g., a flexible spacer or a PEG In an ment, the first member comprises a sequence that is complementary to the sequence of an oligonucleotide ed to a capture substrate.
In an embodiment, the second member comprises (e.g., from 5’ to 3’) one, two, or all of: (i) a sequence that is complementary to at least a portion of the first member; (ii) a universal priming sequence (e.g., for PCR amplification or next-generation sequencing); and (iii) a sequence complementary to a target sequence, e.g., an HCVRS and/or an LCVRS. In an embodiment, the universal priming sequence is identical, or substantially identical, to the sequence that is complementary to at least a portion of the first member. In another embodiment, the universal priming sequence is different from the sequence that is mentary to at least a portion of the first member. In an embodiment, the second member ses a sequence for homologous recombination (e.g., in a yeast or mammalian cell).
In an aspect, the disclosure features an isolated linkage reaction site, e. g., a e micro- chamber, which is an isolated linkage reaction site described herein (e.g., sing a nucleic acid encoding an HCVR and a nucleic acid encoding a LCVR, wherein the HCVR and the LCVR are matched).
In an embodiment, the isolated linkage reaction site, e.g., a linkage micro-chamber, does not include a nucleic acid encoding an HCVR or an LCVR from a different cell.
In an embodiment, the isolated linkage reaction site, e.g., a linkage micro-chamber, comprises a splint oligonucleotide (e.g., a splint oligonucleotide described herein) that is capable of hybridizing to a sequence sing the junction of the HC strand and the LC strand, or a sequence complementary thereof, to form a ed region at the site of on.
In an ment, the isolated linkage reaction site, e. g., a linkage micro-chamber, comprises a reagent that can covalently link nucleic acids, e.g., a ligase, e.g., a thermostable ligase, In an aspect, the disclosure features a method of making a nucleic acid sequence comprising a sequence that encodes an a chain t (AC element) of a TCR 0L chain le region (ACVR) and a [3 chain element (BC element) of a TCR [3 chain variable region (BCVR), and wherein the ACVR and the BCVR are matched, the method comprising: a) acquiring an ed production reaction site, e.g., a production chamber, comprising: i) an a chain (AC) strand, wherein the AC strand is a strand of an or chain double-stranded cDNA (AC ds cDNA) comprising a t that encodes an AC element of the ACVR from a cell, e.g., an or chain variable region sequence (ACVRS); and ii) a [5 chain (BC) strand, n the BC strand is a strand of a [3 chain ds cDNA (BC ds cDNA) comprising a t that encodes a BC element of the BCVR from the cell, e.g., a [3 chain variable region sequence (BCVRS), and b) covalent linking, e. g., ligation, of the first strand to the second strand, n the isolated production reaction site, e.g., a production chamber, does not include a nucleic acid encoding an ACVR or a BCVR from a cell other than the cell (e.g., a different cell, e.g., a different T cell), thereby making a nucleic acid sequence sing a sequence that encodes an AC element of an ACVR and a BC element of a BCVR, wherein the ACVR and the BCVR are matched.
In an embodiment, the AC element comprises, or consists of, an ACVRS, or a functional fragment thereof (e.g., an antigen binding fragment thereof). In an embodiment, the BC element comprises, or consists of, a BCVRS, or a functional fragment thereof (e.g., an antigen binding fragment thereof).
In an embodiment, the AC ds cDNA comprises a segment that s an ACVRS. In an embodiment, the BC ds cDNA comprises a segment that encodes a BCVRS. In an embodiment, the AC ds cDNA comprises a segment that encodes an ACVRS, and the BC ds cDNA comprises a segment that encodes a BCVRS.
In an embodiment, the cell is an immune cell, e.g., a T cell, 6.3., a human T cell. In an embodiment, the cell is a mammalian cell or an avian cell.
In an embodiment, the nucleic acid sequence is configured such that, when expressed, the AC t and the BC element (e.g., the ACVRS and the BCVRS) form a functional antigen binding molecule, e.g., a single chain or a complex of a TCR or chain and a [3 chain. In an embodiment, the antigen binding molecule, e.g., a TCR a chain andior a [3 chain, is functional in vitro, ex vivo, or in vivo, e.g., as determined by a method or assay described herein.
In an embodiment, acquiring an isolated production reaction site, e.g., a production micro- chamber, comprises: a) acquiring a capture substrate bound to: (i) a first double-stranded cDNA (ds cDNA) comprising a strand that is complementary to a first mRNA that encodes an ACVR from a cell; and (ii) a second ds cDNA comprising a strand complementary to a second mRNA encoding a BCVR from the cell (the cDNA loaded capture substrate), and b) maintaining the isolated production reaction site, e.g., the tion chamber, under conditions that allow amplification of the first and second ds cDNAs, to produce: a plurality of AC ds cDNAs comprising a segment that encodes an AC element of the ACVR from the cell, e. g., an ACVRS; and a plurality of BC ds cDNAs comprising a segment that encodes a BC element of the BCVR from the cell, e.g., a BCVRS.
In an embodiment, the AC ds cDNA is cal, or substantially cal, to the first ds cDNA. For example, the sense strand of the AC ds cDNA is at least 80%, 85 %, 90%, 95%, 98%, 99%, or 100% identical to, or differs by no more than 1, 2, 5, 10, 15, 20, 25, 30, 35, 40, 45, or 50 nucleotides from, the sense strand of the first ds cDNA, and/or the antisense strand of the AC ds cDNA is at least 80%, 85%, 90%, 95%, 98%, 99%, or 100% identical to, or s by no more than 1, 2, 5, 10, 15, 20, 25, 30, 35, 40, 45, or 50 nucleotides from, the antisense strand of the first ds cDNA.
In an embodiment, the BC ds cDNA is identical, or ntially identical, to the second ds cDNA. For example, the sense strand of the BC ds cDNA is at least 80%, 85%, 90%, 95%, 98%, 99%, or 100% identical to, or differs by no more than 1, 2, 5, 10, 15, 20, 25, 30, 35, 40, 45, or 50 nucleotides from, the sense strand of the second ds cDNA, and/or the antisense strand of the BC ds cDNA is at least 80%, 85%, 90%, 95%, 98%, 99%, or 100% identical to, or differs by no more than 1, 2, 5, 10, 15, 20, 25, 30, 35, 40, 45 , or 50 nucleotides from, the antisense strand of the second ds cDNA.
In an embodiment, the AC strand is a sense strand. In an embodiment, the BC strand is a sense strand. In an embodiment, the AC strand is an antisense strand. In an embodiment, the BC strand is an antisense strand. In an ment, both the AC strand and the BC strand are sense strands. In an ment, both the AC strand and the BC strand are nse strands.
In an embodiment, the capture ate comprises a bead, e.g., a magnetic bead. In an embodiment, the capture substrate comprises a moiety (e.g., an oligonucleotide) which binds to cDNA, e.g., (i) a moiety which binds to the AC strand; (ii) a moiety which binds to the BC strand; or (iii) both (i) and (ii). In an embodiment, the moiety which binds to the AC strand is different from the moiety which binds to the BC strand, e.g., to facilitate creating conditions favorable to ing similar levels of each DNA molecule type. In an embodiment, the moiety which binds to the AC strand is identical to the moiety which binds to the BC strand.
In an embodiment, the first mRNA and the second mRNA are disposed on an mRNA loaded capture substrate.
In an embodiment, the isolated production reaction site, e.g., the production micro-chamber, comprises: a reagent mixture suitable for producing, from the first and second mRNAs (e.g., after the first and second mRNAs are released from the loaded mRNA capture substrate into a solution), a first cDNA comprising a t that encodes an AC element of the ACVR of the cell, e.g., an ACVRS, and a second cDNA comprising a segment that encodes a BC element of the BCVR of the cell, e.g., a BCVRS.
In an ment, the isolated production reaction site, e.g., production micro-chamber, comprises primers that mediate the production of the first ds cDNA. In an embodiment, the isolated production reaction site, e.g., production micro-chamber, comprises primers that mediate the production of the second ds cDNA.
In an embodiment, a cDNA strand that is complementary to a first mRNA that encodes an ACVR from a cell is made by reverse transcription of the first mRNA. In an ment, a cDNA strand that is complementary to a second mRNA that encodes a BCVR from a cell is made by reverse transcription of the second mRNA.
In an embodiment, the reverse transcription takes place in the isolated production reaction site, e.g., a production-micro chamber. In an embodiment, the e transcription takes place in an isolated cell reaction site, e.g., a cell isolation chamber. In an embodiment, the reverse transcription takes place outside the isolated production reaction site, e.g., a production micro- chamber, or outside an isolated cell reaction site, e.g., a cell isolation micro-chamber. In an embodiment, the reverse transcription takes place e the isolated production reaction site, e. g., a production-micro chamber, and outside an isolated cell reaction site, e.g., a cell isolation micro- chamber. In an embodiment, the reverse transcription takes place outside an isolated on site, 6.3., outside a micro-chamber.
In an embodiment, the amplification ses 20 or fewer cycles, e. g., 15 or fewer, 14 or fewer, 13 or fewer, 12 or fewer, 11 or fewer, 10 or fewer, 9 or fewer, 8 or fewer, 7 or fewer, 6 or fewer, or 5 or fewer cycles.
In an embodiment, the reverse transcription and/or amplification uses one or more primers, e.g., comprising a sequence specific for an ACVRS and/or a BCVRS.
In an embodiment, the reverse transcription and/or amplification comprises using two or more primers that mediate the production of the AC ds cDNA, wherein at least one primer ses a nucleotide modification, and wherein at least one primer does not comprise a nucleotide modification.
In an embodiment, the amplification comprises using two or more primers that mediate the production of the BC ds cDNA, n at least one primer comprises a nucleotide modification, and wherein at least one primer does not comprise a nucleotide modification.
In an embodiment, at least one primer comprises a nucleotide modification, e.g., which reduces, e.g., inhibits, DNA synthesis, e.g., by a DNA polymerase. In an embodiment, at least one primer does not comprise a tide modification, e.g., which s, e.g., inhibits, DNA sis, e.g., by a DNA polymerase.
In an embodiment, the nucleotide modification inhibits a DNA polymerase from ing the DNA. Without wishing to be bound by theory, it is believed that in an embodiment, any chemical entity that reduces (e.3., blocks) DNA polymerase extension can be used in accordance with the methods described herein.
In an embodiment, the nucleotide modification is an insertion of a spacer to the primer, e. g., between two adjacent nucleotides in the primer. In an embodiment, the spacer is a e spacer. In an embodiment, the spacer is a carbon spacer (e.g., -(CH2)n-, n n=3, 4, 5, 6, 7, 8, 9, 10, or more), two or more (e.g., three, four, five, six, seven, eight, nine, ten, or more) abasic nucleotides, or a polyethylene glycol (PEG) spacer. In an embodiment, the spacer is a PEG spacer. In an ment, the nucleotide modification is 2’-O-methyl, 2’-OH, 2’-NH2, or uracil, e.g., to a ribose.
In an embodiment, the tide modification is located internally or at the 3’ end of the primer. In an embodiment, at least one primer comprises (i) a first member; (ii) a second ; and optionally (iii) a third member, e.g., sing a nucleotide modification described herein, e.g., located between (i) and (ii).
In an embodiment, the first member is capable of annealing with the second member. In an embodiment, the first member is capable of ing with the second member in the same primer, e.g., through intra-molecular ization, e.g., to form a hairpin structure comprising a duplex region of 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, more basepairs. In r ment, the first member is capable of annealing hybridizing with the second member in a different primer, e. g., through inter-molecular hybridization, e. g., to form a double-stranded structure comprising a duplex region of 4, 5,6, 7, 8, 9, 10, ll, 12, l3, 14, 15, l6, l7, 18, 19, 20, more basepairs. Without wishing to be bound by theory, it is believed that in an embodiment, there are at least two secondary structures that the modified primers can form and facilitate reduction (e.g., prevention) of ition to substrate (e.g., bead) capture. For example, the secondary structure can be a hairpin-like structure formed by molecular hybridization (within the same primer), or the secondary structure can be a duplex structure formed by inter-molecular hybridization (between two different primers).
In an embodiment, the first member comprises a sequence that is complementary to the sequence of an oligonucleotide attached to the capture substrate. In an embodiment, the second member comprises (6.3., from 5’ to 3’) one, two, or all of: (i) a sequence that is complementary to at least a portion of the first member; (ii) a universal priming sequence (e.g., for PCR amplification or next-generation sequencing); and (iii) a sequence complementary to a target sequence, e.g., an ACVRS and/or a BCVRS. In an embodiment, the universal priming sequence is identical, or substantially identical, to the sequence that is complementary to at least a portion of the first member.
In r embodiment, the universal priming sequence is different from the ce that is complementary to at least a portion of the first . In an embodiment, the second member comprises a sequence for homologous recombination (e.g., in a yeast or mammalian cell).
In an embodiment, at least one primer ses a sequence encoding at least a n of a linker sequence, or a complementary sequence thereof. In an embodiment, the primer that comprises a sequence ng at least a portion of a linker sequence, or a complementary sequence thereof, is phosphorylated, e.g., 5’ phosphorylated. Without wishing to be bound by theory, it is believed that in an embodiment, any sequence with the general ties of flexibility (e.g., facilitated by glycine) and hydrophilicity can work effectively in accordance with the methods described herein. Exemplary linkers can generally have overrepresentation of one or more of Gly, Ser, Thr, or Ala and underrepresentation of hydrophobic residues, e.g., one or more of Trp, Tyr, Phe, Cys, Met, Leu, or Ile.
The length of the primer may vary, e.g., 3-50 amino acid residues (e.g., 5-45, 10-40, 15-35, 20-30, 10- 20, 10-30, 20-40, or 30-40 amino acid residues). In an embodiment, the linker sequence comprises, or consists of, ((Gly)m-Ser))n, where m=3, 4, 5, or more and n=1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more. In an ment, the linker ce comprises, or consists of, (Gly-Gly-Gly-Gly-Ser)n, where n=1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more.
In an embodiment, the primer is a primer described herein, e. g., in Examples.
In an embodiment, the reverse transcription, the amplification, or both, occurs in a solution in the isolated production reaction site, e.g., production micro-chamber. In an embodiment, the reverse transcription, the amplification, or both, does not occur on the substrate (e.g., bead). For example, the reverse ription, the amplification, or both, can occur on in a solution within a droplet.
In an embodiment, the AC ds cDNA comprises a 5’ overhang, e. g., a 5’ overhang that is capable of hybridizing to an oligonucleotide attached to a capture substrate. In an embodiment, the AC ds cDNA ses a blunt end, e.g., a blunt end comprising a 5’ phosphate. In an embodiment, the BC ds cDNA comprises a 5’ ng, e.g., a 5’ ng that is capable of hybridizing to an oligonucleotide attached to a e substrate. In an embodiment, the BC ds cDNA comprises a blunt end, e.g., a blunt end comprising a 5’ phosphate. In an embodiment, the AC ds cDNA and the BC ds cDNA comprise sticky ends, e.g., both have 5’ overhangs.
In an embodiment, the AC strand and the BC strand are ntly linked, e.g., ligated, to produce a single stranded nucleic acid sequence, wherein the AC and BC strands are both sense strands or both antisense strands. In an embodiment, a denatured AC strand of the AC ds cDNA to a denatured BC strand of the BC ds cDNA are covalently linked, e.g., ligated, wherein the AC and BC strands are both sense strands or both antisense strands. In an embodiment, the AC strand is present in the AC ds cDNA and the BC strand is present in the BC ds cDNA, and wherein the AC ds cDNA and the BC ds cDNA are covalently linked, e.g., ligated, e.g., to produce a double stranded nucleic acid sequence.
In an embodiment, the covalent linking, e.g., on, occurs in the isolated production reaction site. In an ment, the isolated production reaction site, e.g., a production micro- chamber, or the ed e reaction site, e.g., a linkage micro-chamber, comprises a reagent that is capable of covalently linking, e.g., ligating, the AC and BC strands or the AC and BC ds cDNAs.
In an embodiment, the isolated tion reaction site, e.g., a production micro-chamber comprises an enzyme that covalently couples the AC and BC strands or the AC and BC ds cDNAs. In an embodiment, the enzyme is a ligase, e.g., a thermal stable ligase. In an embodiment, the covalent linking comprises ligase thermocycling.
In an ment, the covalent linking, e. g., on, occurs in a site different from the isolated production reaction site, e.g., occurs in an isolated linkage reaction site, e.g., a linkage micro- chamber. In an embodiment, the AC strand and the BC strand are transferred from the isolated production site to the isolated linkage reaction site, e.g., a linkage micro-chamber, and the covalent linking occurs in the isolated linkage reaction site, e.g., a linkage micro-chamber. In an embodiment, the isolated linkage on site, e.g., a linkage micro-chamber, comprises a t that is e of covalently linking, e.g., ligating, the AC and BC strands or the AC and BC ds cDNAs. In an embodiment, the isolated linkage reaction site, e.g., a linkage micro-chamber, ses an enzyme that covalently couples the AC and BC s or the AC and BC ds cDNAs. In an embodiment, the enzyme is a ligase, e.g., a thermal stable ligase. In an embodiment, the covalent linking ses ligase thermocycling.
In an embodiment, the covalent linking, e.g., ligation, comprises: (a) heating the isolated linkage reaction site, e. g., the linkage micro-chamber, under conditions (e.g., at 950C) that allow denaturation of the AC strand and the BC ; (b) cooling the isolated linkage reaction site, e. g., the linkage micro-chamber, under conditions (e.g., at 50-65°C) that allow hybridization of the splint oligonucleotide to the AC strand and the BC strand; (c) maintaining the isolated linkage reaction site, 6.3., the linkage micro-chamber, under conditions (e.g., at 45-650C) that allow on of the AC strand and the BC strand (e.g., formation of phosphodiester bond between the AC strand and the BC strand); and (d) repeating steps (a), (b), and (c) sequentially for 2, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, or more cycles.
In an embodiment, the AC strand and the BC strand are covalently linked, e.g., ligated, in the presence of a splint ucleotide. In an ment, the splint oligonucleotide is hybridized to a sequence comprising the junction of the AC strand and the BC strand, or a sequence complementary thereof, and forms a duplexed region at the site of ligation. In an embodiment, the splint oligonucleotide comprises a modification (e.g., an NHZ group) that inhibits DNA synthesis, e.g., by a DNA rase. In an ment, the modification is at the 3’ end of the splint oligonucleotide.
In an embodiment, a strand complimentary to the ntly linked, e.g., ligated, AC and BC strands is produced by amplification.
In an embodiment, the method, e.g., the step of covalent linkage, does not include a step of overlap extension polymerase chain on (OE-PCR), also known as splicing by overlap extension or splicing by overhang extension (SOE) PCR.
In an embodiment, the method further comprises, prior to acquiring the isolated production reaction site, e.g., a production micro-chamber, acquiring an mRNA loaded capture substrate.
In an ment, ing the mRNA loaded capture substrate comprising: a) ing an isolated cell reaction site, e.g., a cell isolation micro-chamber, comprising: i) a cell; and ii) a capture substrate capable of binding a first mRNA ng an ACVR from the cell and a second mRNA encoding a BCVR from the cell; and b) maintaining the isolated cell reaction site, e.g., the cell isolation micro-chamber, under ions that allow lysis of the cell and binding of the capture substrate with the first mRNA and the second mRNA to form the mRNA loaded capture substrate, wherein the isolated cell reaction site, e.g., cell isolation micro-chamber, does not include a nucleic acid encoding an ACVR or a BCVR from a cell other than the cell (e.g., a different cell).
In an embodiment, the isolated cell reaction site, e.g., cell isolation micro-chamber, comprises a lysing reagent, e.g., a detergent. In an embodiment, the cell is lysed by heat or an enzyme. In an embodiment, the capture substrate comprises a moiety (e.g., an oligonucleotide) which binds mRNA, e.g., an dT).
In an embodiment, the method further comprises releasing the mRNA loaded capture substrate from the isolated cell reaction site, e.g., the cell isolation micro-chamber. In an ment, the releasing step is med in the presence of a A) or poly(dT) oligonucleotide, e.g., to reduce cross-binding of non-captured mRNA.
In an embodiment, the mRNA loaded capture substrate is transferred from the isolated cell reaction site, e.g., the cell isolation micro-chamber, to the isolated production reaction site, e.g., the production micro-chamber.
In an ment, the method further comprises releasing the nucleic acid sequence from the isolated production reaction site, e.g., the tion micro-chamber. In an embodiment, the method further comprises amplifying the nucleic acid sequence. In an embodiment, amplification of the nucleic acid sequence occurs outside the isolated production reaction site, e.g., the production micro- chamber, e. g., after the nucleic acid is released from the ed tion reaction site, e.g., the production micro-chamber. In an embodiment, amplification of the nucleic acid ce occurs at the ed production reaction site, e.g., the production micro-chamber.
In an embodiment, the method further comprises sequencing all or a portion of the c acid sequence.
In an embodiment, the method further comprises inserting all or a portion of nucleic acid sequence into a vector. In an embodiment, the vector supplies an additional AC element or BC element not included in the nucleic acid sequence. In an embodiment, the method further comprises expressing the vector.
In an embodiment, the method further comprises expressing the nucleic acid sequence to e a polypeptide comprising a segment that s an AC element of the ACVR, e. g., an ACVRS, and a segment that encodes a BC element of the BCVR, e.g., a BCVRS. In an embodiment, the BC element is N—terminal to the AC element in the polypeptide. In an embodiment, the AC element is C-terminal to the BC element in the polypeptide.
In an ment, the method further comprises contacting the polypeptide with an antigen.
In an embodiment, the method r comprises determining if the polypeptide binds the antigen, in vitro, ex vivo, or in viva, e.g., by a method or assay bed herein.
In an embodiment, the disclosure features a method of making a nucleic acid sequence comprising a sequence that encodes a TCR 0L chain element (AC element) of TCR 0L chain variable region (ACVR) and a TCR [3 chain element (BC element) of a TCR [3 chain variable region (BCVR), and wherein the ACVR and BCVR are matched, comprising: a) acquiring an isolated cell on site (e.g., an isolated cell reaction site described herein), e.g., a cell isolation micro-chamber, comprising: i) a cell (e.g., a cell described herein); and ii) a capture substrate (e.g., a capture substrate described herein) capable of binding a first mRNA encoding an ACVR from the cell and a second mRNA encoding a BCVR from the cell; b) maintaining the isolated cell reaction site, e.g., the cell isolation micro-chamber, under conditions that allow lysis of the cell and binding of the capture substrate with the first mRNA and the second mRNA to form an mRNA loaded e substrate, wherein the isolated cell reaction site, e.g., cell isolation micro-chamber, does not e a nucleic acid encoding an ACVR or a BCVR from a cell other than the cell (e.g., a different cell); c) contacting the mRNA loaded capture substrate with a reaction mixture, e.g., a reaction mixture comprising e transcriptase, that uses the loaded mRNA as a template to make cDNA (this can occur, e.g., in the isolated cell on site, in an isolated tion reaction site, or in neither, e.g., not in an isolated reaction site); d) acquiring an isolated production reaction site (e. g., an isolated production on site described herein), e.g., a production micro-chamber, comprising: i) a TCR 0L chain (AC) strand, wherein the AC strand is a strand of a TCR a chain double-stranded cDNA (AC ds cDNA) comprising a segment that encodes an AC element of the ACVR from the cell, e.g., a TCR 0L chain variable region sequence (ACVRS); and ii) a TCR [5 chain (BC) , wherein the BC strand is a strand of a TCR [3 chain double-stranded cDNA (BC ds cDNA) comprising a segment that encodes a BC element of the BCVR from the cell, e. g., a TCR [3 chain variable region sequence (BCVRS), wherein the isolated production reaction site, e.g., a production micro-chamber, does not include a nucleic acid encoding an ACVR or a BCVR from a cell other than the cell (e.g., a different cell); and e) covalent linking, e.g., ligation, of the AC strand to the BC .
In an embodiment, one or more (e.g., two, three, four, or all) of the steps a)-e) are performed in accordance with a method bed herein. In an embodiment, each of the steps a)-e) is performed in accordance with a method bed herein.
In an aspect, the disclosure features a method of making a nucleic acid sequence comprising a sequence that encodes a TCR 0c chain element (AC t) of a TCR a chain variable region (ACVR) and a TCR B chain element (BC element) of a TCR [3 chain variable region (BCVR), and wherein the ACVR and BCVR are matched, comprising: a) acquiring an isolated cell reaction site (e.g., an isolated cell on site described herein), e.g., a cell ion chamber, comprising: i) a cell (e.g., a cell described herein); and ii) a capture substrate (e. g., a capture substrate described herein) capable of binding a first mRNA encoding an ACVR from the cell and a second mRNA encoding a BCVR from the cell; b) maintaining the isolated cell reaction site, e.g., the cell ion micro-chamber, under conditions that allow lysis of the cell and binding of the capture substrate with the first mRNA and the second mRNA to form the mRNA loaded capture substrate, wherein the isolated cell reaction site, e.g., cell isolation micro-chamber, does not include a nucleic acid encoding an ACVR or a BCVR from a cell other than the cell (e.g., a different cell); c) ing an isolated production reaction site (e.g., an isolated production reaction site described ), e.g., a production micro-chamber, ses: contacting the mRNA loaded capture substrate with a reaction mixture, e.g., a reaction mixture comprising e transcriptase, that uses the loaded mRNA as a te, to produce: a first double-stranded cDNA (ds cDNA) comprising a strand that is complementary to a first mRNA that encodes an ACVR from a cell; and a second ds cDNA comprising a strand complementary to a second mRNA encoding a BCVR from the cell (the cDNA loaded e substrate); wherein the isolated production reaction site, e.g., a production micro-chamber, does not include a nucleic acid encoding an ACVR or a BCVR from a cell other than the cell (e. g., a different cell). d) maintaining the isolated production reaction site, e.g., the production micro-chamber, under ions that allow amplification of the first and second ds cDNAs, to produce: a plurality of AC ds cDNAs comprising a segment that s an AC element of the ACVR from the cell, e.g., an ACVRS; and a plurality of BC ds cDNAs comprising a segment that encodes a BC element of the BCVR from the cell, e.g., a BCVRS; e) acquiring an isolated linkage reaction site (e.g., an ed linkage reaction site described herein), e.g., a linkage micro-chamber, comprising: covalent linking, e. g., ligation, of a strand of the AC ds cDNA (AC strand) to a strand of the BC ds cDNA (BC strand), n the AC and BC strands are both sense strands or antisense strands; and f) amplifying the covalently linked, e.g., ligated, AC and BC strands.
In an embodiment, one or more (e. g., two, three, four, five, or all) of the steps a)-f) are performed in accordance with a method described herein. In an embodiment, each of the steps a)-f) is performed in accordance with a method described herein.
In an aspect, the disclosure features a method of making a library comprising a plurality of unique members, the method comprising: making the plurality of members, wherein each of the members comprises a sequence that encodes a TCR a chain element (AC element) of a TCR a chain variable region (ACVR) and a TCR [3 chain element (BC t) of a TCR [3 chain le region (BCVR), and wherein the ACVR and BCVR are matched, made by a method described , wherein each unique nucleic acid sequence of the plurality comprises an AC t and a BC element from a different unique cell (e. g., a cell described herein), thereby making a library comprising a plurality of unique members.
In an embodiment, the plurality of unique members ses at least 104, 105, 106, 107, 108, or 109 unique s. In an embodiment, the plurality of unique members ses 104 to 109, 104 to 108, 104 to 107, 104 to 106, 104 to 105, 108 to 109, 107 to 109, 106 to 109, 105 to 109, 105 to 10", 106 to 107, 104 to 105, 105 to 106, 106 to 107, 107 to 108, or 108 to 109 unique s. In an embodiment, at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%, of the members in the library are unique members (which encode d AC element and BC element sequences). In an embodiment, less than 20%, 15%, 10%, 5%, 4%, 3%, 2%, or 1%, of the members in the library are unique members (which encode d AC element and BC element sequences).
In an aspect, the disclosure features a y comprising a plurality of unique members, wherein, i) each unique member of the plurality comprises a segment that encodes an AC element, e.g., an ACVRS, and a segment that encodes a BC element, e.g., a BCVRS, wherein the AC element and the BC element in each unique member is matched; ii) each unique member of the plurality comprises a segment that encodes an AC element, e.g., an ACVRS, and a segment that encodes a BC t, e.g., a BCVRS, from a different unique cell; and iii) the library comprises one or more (e.g., two, three, four, or all) of the following properties: a) the library is made by a method described herein; b) the plurality of unique members comprises at least 104, 105, 106, 107, 108, or 109 unique nucleic acid sequences; c) the plurality of unique members comprises 104 to 109, 104 to 108, 104 to 107, 104 to 106, 104 to 105, 108 to 109, 107 to 109, 106 to 109, 105 to 109, 105 to 103, 106 to 107, 104 to 105, 105 to 106, 106 to 107, 107 to 108, or 108 to 109 unique members; d) at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%, of the members in the library are unique members (which encode matched AC element and BC element sequences); or e) less than 20%, 15%, 10%, 5%, 4%, 3%, 2%, or 1%, of the members in the library are unique members (which encode matched AC element and BC element sequences).
In an embodiment, each unique member of the plurality is configured such that, when expressed, the AC element, e.g., the ACVRS, and the BC element, e.g., the BCVRS, form a functional antigen binding molecule, e.g., a single chain or a x of a TCR a chain and a [3 chain.
In an embodiment, the library is a display library. In an embodiment, each of the members of the plurality further encodes a ptide that results in display of the member on the surface of a display entity. In an embodiment, the library is a phage display library. In an embodiment, the library is a yeast display library. In an embodiment, the library is a mammalian display library.
In an aspect, the disclosure es a method of making a binding ptide (e.g., a polypeptide sing an AC element and a BC element), the method comprising: a) acquiring a library described herein, e.g., by a method described herein; and b) expressing a polypeptide encoded by a unique nucleic acid of the library.
In an embodiment, the method further comprises contacting the polypeptide with an antigen.
In an embodiment, the method r comprises retrieving (e.g., isolating or purifying) the c acid that encodes a polypeptide that binds the antigen.
In an aspect, the disclosure features an isolated production reaction site, e.g., a production micro-chamber, which is an isolated production reaction site described herein (e.g., comprising a nucleic acid encoding an ACVR and a nucleic acid encoding a BCVR, wherein the ACVR and the BCVR are matched).
In an embodiment, the isolated production reaction site, e.g., a production micro-chamber, does not include a nucleic acid ng an ACVR or a BCVR from a ent cell.
In an embodiment, the isolated production on site, e.g., a production micro-chamber, comprises one, two, or all of: (i) one or more primers specific to V gene sequences of the AC and BC; (ii) one or more primers specific to overhangs introduced onto the AC and BC cDNAs; or (iii) one or more primers comprising a first , a second member, and a third member comprising a nucleotide modification (e.g., a spacer) located between the first and second s, n the first member is capable of annealing with the second member of the same primer or a different primer, e.g., forming a structure comprising a duplex region of 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, more basepairs.
In an embodiment, the isolated production reaction site, e.g., a production micro-chamber, does not comprise a reagent that can ntly link nucleic acids, e.g., a ligase, e.g., a thermostable 1i gase. In another embodiment, the isolated production reaction site, e. g., a tion micro- chamber, comprises a reagent that can covalently link nucleic acids, e.g., a ligase, e.g., a thermostable ligase.
In an aspect, the disclosure features a self-annealing oligonucleotide comprising a first member, a second , and third member comprising a nucleotide modification (e. g., a spacer) located between the first and second members, wherein the first member is capable of annealing with the second member of the same oligonucleotide (e.g., for a method of making a nucleic acid sequence comprising a sequence that encodes an AC element of an ACVR and a BC element of a BCVR, wherein the ACVR and BCVR are matched).
In an embodiment, the first and second members are e of forming a n structure comprising a duplex region of 4, 5,6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, more basepairs. In an ment, the first member is 5-40 nucleotides, e.g., 5-10, 5-20, 5-30, 30-40, 20- 40, 10-30, 10-30, or 15-25 nucleotides, in length. In an embodiment, the second member is 5-40 nucleotides, e.g., 5-10, 5-20, 5-30, 30-40, 20-40, 10-30, 10-30, or 15-25 nucleotides, in length.
In an embodiment, the spacer is a spacer described , e.g., a flexible spacer or a PEG spacer.
In an embodiment, the first member comprises a sequence that is complementary to the sequence of an ucleotide attached to a capture substrate.
In an embodiment, the second member comprises (e.g., from 5’ to 3’) one, two, or all of: (i) a sequence that is complementary to at least a portion of the first member; (ii) a universal priming sequence (e.g., for PCR amplification or next-generation sequencing); and (iii) a sequence complementary to a target sequence, e.g., an ACVRS and/or a BCVRS. In an embodiment, the universal priming sequence is identical, or substantially identical, to the sequence that is complementary to at least a portion of the first member. In r embodiment, the universal g sequence is different from the sequence that is complementary to at least a portion of the first member. In an embodiment, the second member comprises a sequence for homologous recombination (e.g., in a yeast or mammalian cell).
In an aspect, the disclosure features an isolated linkage reaction site, e.g., a linkage micro- chamber, which is an isolated linkage reaction site described herein (e.g., comprising a nucleic acid encoding an ACVR and a nucleic acid encoding a BCVR, wherein the ACVR and the BCVR are d).
In an embodiment, the isolated linkage on site, e.g., a linkage chamber, does not include a nucleic acid encoding an ACVR or a BCVR from a different cell.
In an embodiment, the isolated linkage reaction site, 6. g., a linkage micro-chamber, comprises a splint oligonucleotide (e.g., a splint oligonucleotide described herein) that is capable of izing to a sequence comprising the junction of the AC strand and the BC strand, or a ce complementary thereof, to form a duplexed region at the site of ligation.
WO 19402 In an embodiment, the isolated linkage on site, e.g., a linkage micro-chamber, ses a reagent that can covalently link nucleic acids, e.g., a ligase, e.g., a thermostable ligase.
In an aspect, the disclosure features a method of making a nucleic acid sequence comprising a sequence that encodes a 7 chain element (GC element) of a TCR V chain variable region (GCVR) and a 5 chain element (DC element) of a TCR 5 chain variable region , and n the GCVR and the DCVR are matched, the method comprising: a) acquiring an isolated tion reaction site, e.g., a production micro-chamber, comprising: i) an y chain (GC) strand, n the GC strand is a strand of an 7 chain double-stranded cDNA (GC ds cDNA) comprising a segment that encodes a GC element of the GCVR from a cell, e.g., an 7 chain variable region sequence (GCVRS); and ii) a 5 chain (DC) strand, wherein the DC strand is a strand of a 5 chain ds cDNA (DC ds cDNA) comprising a segment that encodes a DC element of the DCVR from the cell, e. g., a 5 chain variable region sequence (DCVRS), and b) covalent linking, e.g., ligation, of the first strand to the second strand, n the isolated production reaction site, e.g., a production micro-chamber, does not include a nucleic acid encoding a GCVR or a DCVR from a cell other than the cell (e.g., a different cell, e.g., a different T cell), y making a nucleic acid sequence comprising a sequence that encodes a GC element of a GCVR and a DC element of a DCVR, wherein the GCVR and the DCVR are matched.
In an embodiment, the GC element comprises, or consists of, a GCVRS, or a functional fragment thereof (e.g., an antigen binding fragment thereof). In an embodiment, the DC element ses, or consists of, a DCVRS, or a functional fragment thereof (e.g., an antigen binding fragment thereof).
In an embodiment, the GC ds cDNA comprises a t that encodes a GCVRS. In an ment, the DC ds cDNA comprises a segment that encodes a DCVRS. In an embodiment, the GC ds cDNA comprises a segment that encodes a GCVRS, and the DC ds cDNA comprises a segment that encodes a DCVRS.
In an embodiment, the cell is an immune cell, e.g., a T cell, 6.3., a human T cell. In an embodiment, the cell is a mammalian cell or an avian cell.
In an embodiment, the nucleic acid sequence is configured such that, when expressed, the GC element and the DC element (e.g., the GCVRS and the DCVRS) form a functional antigen binding molecule, e.g., a single chain or a complex of a TCR 7 chain and a 5 chain. In an embodiment, the antigen binding molecule, e.g., a TCR V chain and/or a 5 chain, is functional in vitro, ex vivo, or in viva, e.g., as determined by a method or assay bed herein.
In an embodiment, ing an isolated production reaction site, e.g., a tion micro- chamber, comprises: a) acquiring a capture substrate bound to: (i) a first double-stranded cDNA (ds cDNA) comprising a strand that is complementary to a first mRNA that encodes a GCVR from a cell; and (ii) a second ds cDNA comprising a strand complementary to a second mRNA encoding a DCVR from the cell (the cDNA loaded capture substrate), and b) maintaining the isolated production reaction site, e.g., the production micro-chamber, under conditions that allow cation of the first and second ds cDNAs, to produce: a plurality of GC ds cDNAs comprising a segment that encodes a GC element of the GCVR from the cell, e.g., a GCVRS; and a plurality of DC ds cDNAs comprising a segment that s a DC t of the DCVR from the cell, e.g., a DCVRS.
In an embodiment, the GC ds cDNA is identical, or substantially identical, to the first ds cDNA. For e, the sense strand of the GC ds cDNA is at least 80%, 85 %, 90%, 95%, 98%, 99%, or 100% identical to, or differs by no more than 1, 2, 5, 10, 15, 20, 25, 30, 35, 40, 45, or 50 nucleotides from, the sense strand of the first ds cDNA, and/or the antisense strand of the GC ds cDNA is at least 80%, 85%, 90%, 95%, 98%, 99%, or 100% identical to, or differs by no more than 1, 2, 5, 10, 15, 20, 25, 30, 35, 40, 45, or 50 tides from, the antisense strand of the first ds cDNA.
In an embodiment, the DC ds cDNA is identical, or substantially identical, to the second ds cDNA. For example, the sense strand of the DC ds cDNA is at least 80%, 85 %, 90%, 95%, 98%, 99%, or 100% identical to, or differs by no more than 1, 2, 5, 10, 15, 20, 25, 30, 35, 40, 45, or 50 nucleotides from, the sense strand of the second ds cDNA, and/or the antisense strand of the DC ds cDNA is at least 80%, 85%, 90%, 95%, 98%, 99%, or 100% identical to, or differs by no more than 1, 2, 5, 10, 15, 20, 25, 30, 35, 40, 45 , or 50 nucleotides from, the antisense strand of the second ds cDNA.
In an embodiment, the GC strand is a sense strand. In an embodiment, the DC strand is a sense strand. In an embodiment, the GC strand is an antisense strand. In an embodiment, the DC strand is an antisense strand. In an ment, both the GC strand and the DC strand are sense strands. In an embodiment, both the GC strand and the DC strand are antisense s.
In an embodiment, the capture substrate ses a bead, e.g., a magnetic bead. In an embodiment, the capture substrate comprises a moiety (e.g., an oligonucleotide) which binds to cDNA, e.g., (i) a moiety which binds to the GC strand; (ii) a moiety which binds to the DC strand; or (iii) both (i) and (ii). In an embodiment, the moiety which binds to the GC strand is different from the moiety which binds to the DC strand, e.g., to facilitate creating conditions favorable to capturing similar levels of each DNA molecule type. In an embodiment, the moiety which binds to the GC strand is identical to the moiety which binds to the DC strand.
In an embodiment, the first mRNA and the second mRNA are disposed on an mRNA loaded capture substrate.
In an embodiment, the isolated production on site, e. g., the production micro-chamber, comprises: a reagent e suitable for producing, from the first and second mRNAs (e.g., after the first and second mRNAs are released from the mRNA loaded capture substrate into a solution), a first cDNA comprising a segment that encodes a GC element of the GCVR of the cell, e.g., a GCVRS, and a second cDNA sing a segment that encodes a DC element of the DCVR of the cell, e.g., a DCVRS.
In an embodiment, the isolated production reaction site, e.g., production micro-chamber, comprises primers that mediate the production of the first ds cDNA. In an embodiment, the isolated production reaction site, e.g., production micro-chamber, comprises primers that mediate the production of the second ds cDNA.
In an embodiment, a cDNA strand that is complementary to a first mRNA that encodes a GCVR from a cell is made by reverse transcription of the first mRNA. In an embodiment, a cDNA strand that is mentary to a second mRNA that encodes a DCVR from a cell is made by reverse transcription of the second mRNA.
In an embodiment, the reverse transcription takes place in the isolated production reaction site, e.g., a production-micro chamber. In an embodiment, the reverse transcription takes place in an isolated cell on site, e.g., a cell isolation micro-chamber. In an embodiment, the reverse transcription takes place outside the isolated production reaction site, e.g., a tion micro- chamber, or outside an isolated cell on site, e.g., a cell isolation micro-chamber. In an embodiment, the reverse transcription takes place outside the isolated production reaction site, e.g., a production-micro chamber, and outside an isolated cell reaction site, e.g., a cell ion micro- chamber. In an ment, the reverse transcription takes place outside an isolated reaction site, e.g., e a micro-chamber.
In an ment, the amplification comprises 20 or fewer cycles, e.g., 15 or fewer, 14 or fewer, 13 or fewer, 12 or fewer, 11 or fewer, 10 or fewer, 9 or fewer, 8 or fewer, 7 or fewer, 6 or fewer, or 5 or fewer cycles.
In an ment, the e transcription and/or amplification uses one or more primers, e.g., comprising a sequence specific for a GCVRS and/or a DCVRS.
In an embodiment, the e transcription and/or amplification comprises using two or more primers that mediate the production of the GC ds cDNA, wherein at least one primer comprises a nucleotide modification, and wherein at least one primer does not comprise a nucleotide modification.
In an embodiment, the amplification comprises using two or more primers that mediate the production of the DC ds cDNA, wherein at least one primer comprises a nucleotide modification, and wherein at least one primer does not comprise a nucleotide modification.
In an embodiment, at least one primer comprises a nucleotide modification, e. g., which reduces, e.g., inhibits, DNA synthesis, e.g., by a DNA polymerase. In an embodiment, at least one primer does not comprise a nucleotide modification, e.g., which reduces, e.g., inhibits, DNA sis, e.g., by a DNA polymerase.
In an embodiment, the nucleotide modification inhibits a DNA polymerase from extending the DNA. Without wishing to be bound by theory, it is ed that in an embodiment, any chemical entity that reduces (e.g., blocks) DNA rase extension can be used in accordance with the methods described herein.
In an embodiment, the nucleotide modification is an insertion of a spacer to the primer, e.g., between two adjacent nucleotides in the primer. In an embodiment, the spacer is a flexible spacer. In an embodiment, the spacer is a carbon spacer (e.g., -(CH2)n-, wherein n=3, 4, 5, 6, 7, 8, 9, 10, or more), two or more (e.g., three, four, five, six, seven, eight, nine, ten, or more) abasic nucleotides, or a polyethylene glycol (PEG) spacer. In an embodiment, the spacer is a PEG spacer. In an embodiment, the tide modification is ethyl, 2’-OH, 2’-NH2, or uracil, e.g., to a ribose.
In an embodiment, the tide modification is located internally or at the 3’ end of the primer. In an ment, at least one primer comprises (i) a first member; (ii) a second member; and optionally (iii) a third member, e.g., comprising a nucleotide modification described herein, e.g., located between (i) and (ii).
In an embodiment, the first member is capable of annealing with the second member. In an embodiment, the first member is capable of annealing with the second member in the same primer, e.g., through intra-molecular hybridization, e.g., to form a hairpin structure comprising a duplex region of 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, more basepairs. In another embodiment, the first member is capable of annealing hybridizing with the second member in a different , e.g., through inter-molecular hybridization, e.g., to form a double-stranded ure comprising a duplex region of 4, 5,6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, more basepairs. t wishing to be bound by theory, it is believed that in an embodiment, there are at least two secondary structures that the modified primers can form and facilitate reduction (e.g., prevention) of ition to substrate (e.g., bead) capture. For example, the secondary structure can be a hairpin-like structure formed by intra-molecular hybridization (within the same primer), or the secondary structure can be a duplex structure formed by inter-molecular hybridization (between two different primers).
In an embodiment, the first member comprises a sequence that is complementary to the sequence of an oligonucleotide attached to the capture substrate. In an embodiment, the second member ses (e.g., from 5’ to 3’) one, two, or all of: (i) a sequence that is complementary to at least a portion of the first member; (ii) a universal priming sequence (e.g., for PCR amplification or next-generation sequencing); and (iii) a sequence complementary to a target sequence, e.g., a GCVRS andior a DCVRS. In an embodiment, the universal priming ce is identical, or substantially cal, to the sequence that is complementary to at least a portion of the first . In another ment, the universal g sequence is different from the sequence that is complementary to at least a portion of the first member. In an embodiment, the second member comprises a sequence for homologous recombination (e.g., in a yeast or mammalian cell).
In an embodiment, at least one primer comprises a sequence encoding at least a portion of a linker sequence, or a complementary ce f. In an embodiment, the primer that comprises a sequence encoding at least a portion of a linker sequence, or a complementary sequence thereof, is phosphorylated, e.g., 5’ phosphorylated. Without wishing to be bound by theory, it is believed that in an embodiment, any sequence with the general properties of flexibility (e.g., facilitated by glycine) and hydrophilicity can work effectively in accordance with the methods described . ary linkers can generally have overrepresentation of one or more of Gly, Ser, Thr, or Ala and underrepresentation of hydrophobic residues, e.g., one or more of Trp, Tyr, Phe, Cys, Met, Leu, or 116.
The length of the primer may vary, e.g., 3-50 amino acid residues (e.g., 5-45, 10-40, 15-35, 20-30, 10- , 10-30, 20-40, or 30-40 amino acid residues). In an embodiment, the linker sequence comprises, or consists of, ((Gly)m-Ser))n, where m=3, 4, 5, or more and n=1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more. In an embodiment, the linker sequence ses, or consists of, (Gly-Gly-Gly-Gly-Ser)n, where n=1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more.
In an embodiment, the primer is a primer bed herein, e.g., in Examples.
In an embodiment, the reverse transcription, the amplification, or both, occurs in a solution in the isolated production reaction site, e.g., production micro-chamber. In an embodiment, the reverse transcription, the amplification, or both, does not occur on the substrate (e.g., bead). For example, the reverse transcription, the amplification, or both, can occur on in a solution within a droplet.
In an ment, the GC ds cDNA comprises a 5’ overhang, e.g., a 5’ overhang that is capable of hybridizing to an oligonucleotide attached to a e substrate. In an embodiment, the GC ds cDNA comprises a blunt end, e.g., a blunt end comprising a 5’ phosphate. In an embodiment, the DC ds cDNA comprises a 5’ overhang, e.g., a 5’ overhang that is e of hybridizing to an oligonucleotide attached to a capture ate. In an embodiment, the DC ds cDNA comprises a blunt end, e. g., a blunt end comprising a 5’ phosphate. In an embodiment, the GC ds cDNA and the DC ds cDNA comprise sticky ends, e.g., both have 5’ overhangs.
In an embodiment, the GC strand and the DC strand are covalently , e.g., ligated, to produce a single stranded nucleic acid sequence, wherein the GC and DC strands are both sense strands or both antisense s. In an embodiment, a red GC strand of the GC ds cDNA to a denatured DC strand of the DC ds cDNA are covalently linked, e.g., ligated, wherein the GC and DC strands are both sense s or both antisense strands. In an embodiment, the GC strand is present in the GC ds cDNA and the DC strand is present in the DC ds cDNA, and n the GC ds cDNA and the DC ds cDNA are covalently linked, e.g., ligated, e.g., to produce a double stranded nucleic acid sequence.
In an embodiment, the covalent linking, e.g., ligation, occurs in the isolated production reaction site. In an embodiment, the isolated production reaction site, e.g., a production micro- r, or the isolated linkage reaction site, e.g., a linkage micro-chamber, comprises a reagent that is capable of covalently linking, e.g., ligating, the GC and DC strands or the GC and DC ds cDNAs.
In an embodiment, the isolated production reaction site, e.g., a production micro-chamber comprises an enzyme that covalently couples the GC and DC strands or the GC and DC ds cDNAs. In an embodiment, the enzyme is a ligase, e. g., a thermal stable ligase. In an embodiment, the covalent linking comprises ligase cycling.
In an embodiment, the covalent linking, e. 3., ligation, occurs in a site different from the isolated tion reaction site, e.g., occurs in an isolated e reaction site, e.g., a linkage micro- chamber. In an embodiment, the GC strand and the DC strand are transferred from the isolated production site to the isolated linkage reaction site, e.g., a linkage micro-chamber, and the covalent linking occurs in the isolated e reaction site, e.g., a linkage micro-chamber. In an embodiment, the isolated linkage reaction site, e.g., a linkage micro-chamber, comprises a reagent that is capable of covalently g, e.g., ligating, the GC and DC strands or the GC and DC ds cDNAs. In an embodiment, the ed linkage reaction site, e. g., a linkage micro-chamber, ses an enzyme that covalently couples the GC and DC strands or the GC and DC ds cDNAs. In an embodiment, the enzyme is a ligase, e.g., a thermal stable ligase. In an embodiment, the covalent g comprises ligase cycling.
In an embodiment, the covalent linking, e. 3., ligation, comprises: (a) heating the isolated linkage reaction site, e. g., the linkage micro-chamber, under conditions (e.g., at 950C) that allow denaturation of the GC strand and the DC strand; (b) cooling the isolated linkage reaction site, e.g., the linkage micro-chamber, under conditions (e.g., at 50-65°C) that allow hybridization of the splint ucleotide to the GC strand and the DC strand; (c) maintaining the isolated linkage reaction site, e.g., the linkage micro-chamber, under conditions (e.g., at 45-650C) that allow ligation of the GC strand and the DC strand (e.g., formation of phosphodiester bond between the GC strand and the DC ); and (d) repeating steps (a), (b), and (c) sequentially for 2, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, or more cycles.
In an embodiment, the GC strand and the DC strand are covalently linked, e.g., ligated, in the presence of a splint oligonucleotide. In an ment, the splint oligonucleotide is hybridized to a sequence comprising the junction of the GC strand and the DC strand, or a sequence mentary f, and forms a duplexed region at the site of ligation. In an embodiment, the splint oligonucleotide comprises a modification (e.g., an NH2 group) that inhibits DNA synthesis, e.g., by a DNA polymerase. In an embodiment, the ation is at the 3’ end of the splint oligonucleotide.
In an embodiment, a strand complimentary to the covalently linked, e.g., ligated, GC and DC strands is produced by amplification.
In an embodiment, the method, e.g., the step of covalent linkage, does not include a step of p extension polymerase chain reaction (OE-PCR), also known as splicing by p extension or splicing by overhang extension (SOE) PCR.
In an embodiment, the method further comprises, prior to acquiring the isolated production reaction site, e.g., a tion micro-chamber, acquiring an mRNA loaded e substrate.
In an embodiment, ing the mRNA loaded capture substrate comprising: a) acquiring an isolated cell reaction site, e.g., a cell isolation micro-chamber, comprising: i) a cell; and ii) a capture substrate capable of binding a first mRNA encoding a GCVR from the cell and a second mRNA encoding a DCVR from the cell; and b) maintaining the isolated cell reaction site, e.g., the cell isolation micro-chamber, under conditions that allow lysis of the cell and binding of the capture substrate with the first mRNA and the second mRNA to form the mRNA loaded capture substrate, wherein the isolated cell on site, e.g., cell isolation chamber, does not include a nucleic acid encoding a GCVR or a DCVR from a cell other than the cell (e.g., a different cell).
In an embodiment, the isolated cell reaction site, e.g., cell isolation micro-chamber, comprises a lysing reagent, e.g., a detergent. In an embodiment, the cell is lysed by heat or an enzyme. In an embodiment, the capture substrate comprises a moiety (e.g., an oligonucleotide) which binds mRNA, e. g., an oligo(dT).
In an embodiment, the method further comprises releasing the mRNA loaded capture substrate from the isolated cell reaction site, e.g., the cell isolation micro-chamber. In an embodiment, the releasing step is med in the presence of a A) or poly(dT) oligonucleotide, e.g., to reduce cross-binding of non-captured mRNA.
In an embodiment, the mRNA loaded capture substrate is transferred from the isolated cell reaction site, e. g., the cell ion chamber, to the isolated production on site, e.g., the production micro-chamber.
In an embodiment, the method r comprises releasing the nucleic acid sequence from the isolated production reaction site, e.g., the production micro-chamber. In an embodiment, the method further comprises ying the nucleic acid sequence. In an embodiment, amplification of the nucleic acid sequence occurs outside the isolated production reaction site, e.g., the production micro- chamber, e. g., after the nucleic acid is released from the isolated production reaction site, 6.55., the production micro-chamber. In an ment, amplification of the nucleic acid sequence occurs at the isolated production reaction site, e.g., the production micro-chamber.
In an embodiment, the method further comprises sequencing all or a portion of the nucleic acid sequence.
In an embodiment, the method further comprises inserting all or a portion of nucleic acid ce into a vector. In an embodiment, the vector supplies an additional GC element or DC element not ed in the c acid sequence. In an embodiment, the method further comprises expressing the vector.
In an embodiment, the method further comprises expressing the nucleic acid sequence to produce a ptide comprising a segment that encodes a GC element of the GCVR, e.g., a GCVRS, and a segment that encodes a DC element of the DCVR, e. g., a DCVRS. In an embodiment, the DC element is N-terminal to the GC element in the polypeptide. In an embodiment, the GC element is C-terminal to the DC element in the ptide.
In an embodiment, the method further comprises contacting the ptide with an antigen.
In an embodiment, the method r comprises determining if the polypeptide binds the antigen, in vitro, ex vivo, or in viva, e.g., by a method or assay described herein.
In an embodiment, the disclosure features a method of making a nucleic acid sequence comprising a sequence that encodes a TCR 7 chain element (GC element) of TCR 7 chain variable region (GCVR) and a TCR 5 chain element (DC element) of a TCR 5 chain variable region (DCVR), and n the GCVR and DCVR are matched, comprising: a) acquiring an ed cell on site (e.g., an isolated cell reaction site described herein), e.g., a cell isolation micro-chamber, comprising: i) a cell (e.g., a cell described herein); and ii) a capture substrate (e.g., a capture substrate described herein) capable of g a first mRNA encoding a GCVR from the cell and a second mRNA encoding a DCVR from the cell; b) maintaining the isolated cell reaction site, e.g., the cell isolation micro-chamber, under conditions that allow lysis of the cell and binding of the capture substrate with the first mRNA and the second mRNA to form an mRNA loaded capture substrate, wherein the isolated cell reaction site, e.g., cell isolation chamber, does not include a nucleic acid encoding a GCVR or a DCVR from a cell other than the cell (e.g., a different cell); c) contacting the mRNA loaded capture substrate with a reaction mixture, e.g., a reaction mixture sing reverse transcriptase, that uses the loaded mRNA as a template to make cDNA (this can occur, e.g., in the isolated cell reaction site, in an isolated production on site, or in neither, e.g., not in an isolated reaction site); d) acquiring an isolated production reaction site (e.g., an isolated tion reaction site described herein), e.g., a production micro-chamber, comprising: i) a TCR 7 chain (GC) strand, wherein the GC strand is a strand of a TCR 7 chain double-stranded cDNA (GC ds cDNA) comprising a segment that encodes a GC t of the GCVR from the cell, e.g., a TCR V chain variable region sequence ); and ii) a TCR 5 chain (DC) , wherein the DC strand is a strand of a TCR 5 chain double-stranded cDNA (DC ds cDNA) comprising a segment that encodes a DC element of the DCVR from the cell, e. g., a TCR 5 chain variable region sequence (DCVRS), n the isolated production reaction site, e.g., a production micro-chamber, does not include a nucleic acid encoding a GCVR or a DCVR from a cell other than the cell (e.g., a different cell); and e) covalent linking, e.g., ligation, of the GC strand to the DC strand.
In an embodiment, one or more (e.g., two, three, four, or all) of the steps a)-e) are performed in accordance with a method described herein. In an embodiment, each of the steps a)-e) is performed in accordance with a method described herein.
In an aspect, the disclosure features a method of making a nucleic acid sequence comprising a sequence that encodes a TCR V chain element (GC element) of a TCR 7 chain variable region (GCVR) and a TCR 5 chain element (DC element) of a TCR 5 chain variable region (DCVR), and wherein the GCVR and DCVR are d, comprising: a) acquiring an isolated cell reaction site (e.g., an isolated cell reaction site described herein), e.g., a cell isolation micro-chamber, comprising: i) a cell (e.g., a cell described herein); and ii) a e ate (e.g., a capture substrate described herein) capable of binding a first mRNA encoding a GCVR from the cell and a second mRNA encoding a DCVR from the cell; b) maintaining the isolated cell reaction site, e.g., the cell isolation micro-chamber, under conditions that allow lysis of the cell and binding of the capture substrate with the first mRNA and the second mRNA to form the mRNA loaded capture substrate, wherein the isolated cell reaction site, e.g., cell isolation micro-chamber, does not include a c acid encoding a GCVR or a DCVR from a cell other than the cell (e.g., a different cell); c) acquiring an isolated production reaction site (e.g., an isolated tion reaction site described herein), e.g., a production micro-chamber, comprises: contacting the mRNA loaded capture substrate with a reaction mixture, e.g., a reaction mixture comprising reverse transcriptase, that uses the loaded mRNA as a template, to produce: a first double-stranded cDNA (ds cDNA) comprising a strand that is complementary to a first mRNA that encodes a GCVR from a cell; and a second ds cDNA comprising a strand complementary to a second mRNA encoding a DCVR from the cell (the cDNA loaded e substrate); wherein the isolated production reaction site, e.g., a production micro-chamber, does not e a c acid encoding a GCVR or a DCVR from a cell other than the cell (e.g., a different cell). d) maintaining the isolated production reaction site, e.g., the tion micro-chamber, under ions that allow amplification of the first and second ds cDNAs, to produce: a plurality of GC ds cDNAs comprising a segment that s a GC element of the GCVR from the cell, e.g., a GCVRS; and a ity of DC ds cDNAs comprising a segment that encodes a DC element of the DCVR from the cell, e.g., a DCVRS; e) acquiring an isolated linkage reaction site (e.g., an isolated linkage reaction site described herein), e.g., a linkage micro-chamber, comprising: covalent linking, e.g., ligation, of a strand of the GC ds cDNA (GC strand) to a strand of the DC ds cDNA (DC strand), wherein the GC and DC strands are both sense strands or nse strands; and f) amplifying the covalently linked, e.g., ligated, GC and DC strands.
In an embodiment, one or more (e.g., two, three, four, five, or all) of the steps a)-f) are med in accordance with a method described herein. In an embodiment, each of the steps a)-f) is performed in accordance with a method described herein.
WO 19402 In an aspect, the disclosure features a method of making a library comprising a plurality of unique members, the method comprising: making the plurality of members, n each of the members comprises a ce that encodes a TCR V chain element (GC element) of a TCR y chain le region (GCVR) and a TCR 6 chain element (DC element) of a TCR 5 chain variable region (DCVR), and wherein the GCVR and DCVR are matched, made by a method bed herein, wherein each unique nucleic acid sequence of the plurality comprises a GC element and a DC element from a different unique cell (e.g., a cell described herein), thereby making a library comprising a plurality of unique members.
In an embodiment, the plurality of unique members comprises at least 104, 105, 106, 107, 108, or 109 unique members. In an embodiment, the plurality of unique members comprises 104 to 109, 104 to 108, 104 to 107, 104 to 106, 104 to 105, 108 to 109, 107 to 109, 106 to 109, 105 to 109, 105 to 108, 106 to 107, 104 to 105, 105 to 106, 106 to 107, 107 to 108, or 108 to 109 unique members. In an embodiment, at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%, of the s in the library are unique members (which encode matched GC element and DC element sequences). In an embodiment, less than 20%, 15%, 10%, 5%, 4%, 3%, 2%, or 1%, of the s in the library are unique members (which encode matched GC element and DC element sequences).
In an aspect, the disclosure features a library comprising a plurality of unique members, wherein, i) each unique member of the ity ses a segment that encodes a GC element, e. g., a GCVRS, and a segment that encodes a DC element, e.g., a DCVRS, wherein the GC element and the DC element in each unique member is matched; ii) each unique member of the plurality comprises a segment that encodes an GC element, e.g., a GCVRS, and a segment that encodes a DC element, e.g., a DCVRS, from a different unique cell; and iii) the library comprises one or more of the ing properties: a) the library is made by a method described ; b) the plurality of unique s comprises at least 104, 105, 106, 107, 108, or 109 unique nucleic acid sequences; c) the plurality of unique members comprises 104 to 109, 104 to 108, 104 to 107, 104 to 106, 104 to 105, 108 to 109, 107 to 109, 106 to 109, 105 to 109, 105 to 103, 106 to 107, 104 to 105, 105 to 106, 106 to 107, 107 to 108, or 108 to 109 unique members; d) at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%, of the members in the library are unique members (which encode matched GC element and DC element sequences); or e) less than 20%, 15%, 10%, 5%, 4%, 3%, 2%, or 1%, of the members in the library are unique members (which encode matched GC element and DC element sequences).
In an embodiment, each unique member of the plurality is configured such that, when expressed, the GC element, e.g., the GCVRS, and the DC element, e.g., the DCVRS, form a functional antigen binding molecule, e.g., a single chain or a complex of a TCR 7 chain and a 5 chain.
In an embodiment, the library is a display y. In an embodiment, each of the members of the plurality further s a polypeptide that results in display of the member on the surface of a y entity. In an embodiment, the y is a phage display library. In an embodiment, the library is a yeast y library. In an embodiment, the library is a mammalian display library.
In an aspect, the disclosure features a method of making a binding ptide (e.g., a polypeptide comprising a GC element and a DC element), the method comprising: a) acquiring a library bed herein, e.g., by a method bed herein; and b) sing a polypeptide encoded by a unique nucleic acid of the library.
In an embodiment, the method further comprises contacting the polypeptide with an n.
In an ment, the method further ses retrieving (e.g., isolating or purifying) the nucleic acid that encodes a polypeptide that binds the antigen.
In an aspect, the disclosure features an isolated production reaction site, e.g., a production micro-chamber, which is an isolated production reaction site described herein (e.g., comprising a nucleic acid encoding a GCVR and a nucleic acid encoding a DCVR, wherein the GCVR and the DCVR are matched).
In an embodiment, the isolated production reaction site, e.g., a production micro-chamber, does not include a nucleic acid encoding a GCVR or a DCVR from a different cell.
In an embodiment, the isolated production reaction site, e.g., a production micro-chamber, comprises one, two, or all of: (i) one or more primers ic to V gene sequences of the GC and DC; (ii) one or more primers c to overhangs introduced onto the GC and DC cDNAs; or (iii) one or more primers comprising a first member, a second member, and a third member comprising a nucleotide modification (e.g., a spacer) located between the first and second members, wherein the first member is capable of annealing with the second member of the same primer or a different primer, e.g., g a structure comprising a duplex region of 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, more basepairs.
In an embodiment, the isolated production reaction site, e.g., a production micro-chamber, does not comprise a reagent that can covalently link nucleic acids, e.g., a , e.g., a thermostable 1i gase. In another embodiment, the isolated production reaction site, e. g., a production micro- chamber, comprises a reagent that can ntly link nucleic acids, e.g., a ligase, e.g., a thermostable ligase.
In an aspect, the disclosure features a self-annealing oligonucleotide comprising a first member, a second , and third member comprising a nucleotide modification (e. g., a spacer) located between the first and second members, wherein the first member is capable of annealing with the second member of the same oligonucleotide (e.g., for a method of making a nucleic acid sequence comprising a sequence that encodes a GC element of a GCVR and a DC element of a DCVR, wherein the GCVR and DCVR are d).
In an embodiment, the first and second members are capable of g a hairpin structure sing a duplex region of 4, 5,6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, more basepairs. In an embodiment, the first member is 5-40 nucleotides, e.g., 5-10, 5-20, 5-30, 30-40, 20- 40, 10-30, 10-30, or 15-25 nucleotides, in length. In an embodiment, the second member is 5-40 nucleotides, e.g., 5-10, 5-20, 5-30, 30-40, 20-40, 10-30, 10-30, or 15-25 nucleotides, in length.
In an embodiment, the spacer is a spacer described , e.g., a flexible spacer or a PEG spacer.
In an embodiment, the first member comprises a sequence that is complementary to the sequence of an oligonucleotide attached to a capture substrate.
In an ment, the second member comprises (e.g., from 5’ to 3’) one, two, or all of: (i) a sequence that is mentary to at least a portion of the first member; (ii) a universal priming sequence (e.g., for PCR amplification or next-generation sequencing); and (iii) a sequence complementary to a target sequence, e.g., a GCVRS and/or a DCVRS. In an embodiment, the universal priming ce is identical, or substantially identical, to the sequence that is complementary to at least a portion of the first member. In another embodiment, the universal priming sequence is different from the ce that is mentary to at least a portion of the first member. In an embodiment, the second member comprises a sequence for homologous recombination (e.g., in a yeast or mammalian cell).
In an aspect, the sure features an isolated linkage reaction site, e.g., a linkage micro- chamber, which is an isolated linkage reaction site described herein (e.g., comprising a nucleic acid encoding a GCVR and a nucleic acid encoding a DCVR, wherein the GCVR and the DCVR are matched).
In an embodiment, the isolated linkage reaction site, e.g., a linkage micro-chamber, does not include a nucleic acid encoding a GCVR or a DCVR from a different cell.
In an ment, the isolated linkage reaction site, 6. g., a linkage micro-chamber, comprises a splint oligonucleotide (e.g., a splint oligonucleotide described herein) that is capable of hybridizing to a sequence sing the junction of the GC strand and the DC strand, or a ce complementary thereof, to form a duplexed region at the site of ligation.
In an embodiment, the isolated linkage reaction site, e.g., a e micro-chamber, comprises a reagent that can covalently link nucleic acids, e.g., a ligase, e.g., a thermostable ligase.
BRIEF DESCRIPTION OF THE DRAWINGS depicts a number of ways of making nucleic acid ce comprising a sequence that encodes a heavy chain element (HC element) of an antibody heavy chain variable region (HCVR) and a light chain element (LC t) of an antibody light chain variable region (LCVR), and wherein the HCVR and LCVR are matched. The A1, B1 and C2 boxes indicate steps occurring in an isolated reaction site, particularly, in an isolated cell reaction site. The C3, D1, D2, D3, D4, D5 and D6 boxes indicate steps occurring in an isolated reaction site, particularly, in an isolated production reaction site. The E1, E2 and E3 boxes indicate steps occurring in an isolated reaction site, ularly, in an ed linkage on site. The C1 box indicates steps that need not occur in an isolated reaction site. As is discussed in the text, the isolated reaction sites are free of nucleic acid that would result in a ched HC and LC element.
FIGS. 2A-2D are a series of diagrams showing an exemplary method of making a nucleic acid comprising a sequence that encodes a heavy chain element (HC element) of an dy heavy chain variable region (HCVR) and a light chain element (LC element) of an antibody light chain variable region (LCVR), and wherein the HCVR and LCVR are matched. In , a cell (e.g., an immune cell, such as a B cell) is lysed and mRNAs encoding an HCVR and a matched LCVR are captured on a bead. In , captured mRNAs are ted to cDNA by reverse transcription followed by amplification by DNA polymerase (PCR) to create cDNA beads comprising matched pairs of HCVR and LCVR cDNAs. A self-annealing primer (e.g., a primer comprising a first member and a second member capable of hybridizing to the first member, with the first and second members separated by a spacer, e.g., a PEG spacer, and further comprising a sequence capable of hybridizing to an HCVR or LCVR sequence) can be used for the reverse transcription on and/or DNA polymerase amplification. In , matched LCVR and HCVR cDNA products can be fused using a ligase cycling reaction, in which matched pairs of LCVRs and HCVRs are brought together using a splint oligo comprising sequences capable of hybridizing to an end of each of the LCVR or HCVR sequences (e.g., the 3’ terminus of the LCVR and the 5’ terminus of the HCVR). In , the fused LCVR/HCVR product can be amplified, e.g., by PCR. is a polyacrylamide gel electrophoresis (PAGE) image g that Taq ligase and Ampligase thermostable ligase (Amp; Lucigen) were capable of efficiently g VH and VL product.
FIGS. 4A-4B are gel electrophoresis images showing that natively paired, linked VH + VL products for each of antibodies 4G2 and 9E10 were successfully produced by ligase cycling. In , denaturing PAGE of ligase g products showed that ligase-containing reactions yielded the linked VH + VL products for each of 4G2 and 9E10, as well as the individual VH and VL polynucleotides. The linked VH + VL ts were not detected in reactions lacking ligase. In FIG.
WO 19402 4B, agarose gel electrophoresis of bulk PCR re-amplification products showed that native pairing was retained for when VH-VL linked polynucleotides for 4G2 and 9E10 were mixed in the PCR reaction.
FIGS. 5A-SB are a graph and diagram showing efficient and specific PCR product capture using a self-annealing primer. In , a series of forward PCR primer designs were tested for their capacity to capture PCR product, ing (1) a VL primer comprising a spacer and with 5’ sequence complementary to oligo on bead and 3’ sequence that is complementary to VL template sequence, (2) a VL primer lacking a spacer and with 5’ sequence complementary to oligo on bead and 3’ sequence that is complementary to VL template sequence, (3) a VL primer lacking 5’ sequence mentary to oligo on bead and 3’ ce that is complementary to VL template sequence, and (4) a VH primer with similar design as in (l) but with 3’-end having sequence complementary to VH template (for DNA polymerase extension). In , the VL primer comprising a spacer was used for ent and specific PCR capture of VL oligo, VH oligo, and VH+VL oligo. Of the remaining primers, only the VH primer was capable of capturing any of the oligos (specifically, the VH oligo and VH+VL oligo). is an agarose gel electrophoresis image showing that natively paired VH-VL products could be produced in drops from nucleic acids ed from cells expressing the 4G2 antibody. NTC = sample in which the entire droplet workflow was performed but no cells were included; PCR NTC = no- template control.
FIGS. 7A-7B are a series of graphs showing that self-annealing primers (in ) can t PCR product capture competition at high levels of unused primer, whereas non-self—annealing primers (in ) can only do so at low levels of unused primer.
DETAILED DESCRIPTION Disclosed herein are polypeptides (e.g., antibody molecules or T cell receptor molecules) that bind to a target molecule or cell, e.g., a human protein or cell, with high affinity and specificity. In an embodiment, the polypeptide is a g polypeptide. In an embodiment, the binding polypeptide is an antibody molecule. In an ment, the binding polypeptide is a TCR molecule (e.g., a soluble TCR molecule). In an embodiment, libraries of the polypeptides, methods for making the polypeptides or libraries, c acid molecules encoding the polypeptides, expression vectors, host cells, and compositions (e.g., pharmaceutical compositions), kits, ners, are also provided. The s described herein are useful for making or selecting functional polypeptides that contain two or more chains that are lly matched or paired. The polypeptides (e.g., antibody molecules or T cell receptor molecules) sed herein can be used (alone or in combination with other agents or therapeutic modalities) to treat, prevent and/or diagnose disorders, such as disorders and conditions disclosed herein.
Without wishing to be bound by theory, it is ed that the methods described herein can facilitate, e. g., high-throughput phenotypic (e.g., binding) screening of ns of B-cellfplasma cell antibodies, and antibody discovery from B-cells derived from different species, including, but not d to, human, mouse, rat, rabbit, or chicken. For example, the only requirement can be dge of primers to appropriately amplify VH and VL sequences from that species.
Since the workflow bed herein is amenable to use in any species, it can significantly improve ability to discover diverse binding polypeptides (e. g., antibodies) to target antigens (post immunization/vaccination), as each species ps different types of binding polypeptides (e.g., antibodies) to an antigen. Immune tolerance issues (e.g., to a target epitope) can be better overcome by using a species which lacks the target antigen or has significant amino acid differences to the target antigen, e.g., chicken has d nce to human antigens/epitopes than human or mouse does to human antigens/epitopes.
The methods described herein can facilitate making a ‘phenotypic copy’ of an dy repertoire in yeast, which are rugged and can be regrown. This tates rigorous and repeated testing of the antibody repertoire, unlike when using primary B-cells, which are sensitive, do no survive long in vitro, and cannot survive rigorous antibody/BCR binding characterizations.
Other methods to generate ly paired VH-VL sequences in droplets can use splicing by overlap extension with DNA polymerase (PCR) to link the DNA, which may have limitations with specificity and can result in heterogeneous products of divergent sizes due to imprecise linking. The ligation methods described here do not suffer from such issues.
Additionally, droplet methods using splicing by overlap extension PCR suffer from an inherent limitation in which any PCR products not fused within drops have the potential to become fused during non-drop PCR amplification due to the common ed sequence between VH and VL. Fusion occurring outside of drops leads to non-native pairing, as chains are not compartmentalized. For the exemplary ligation workflow described herein, there is no need to add common sequence to VH and VL, and therefore this issue is precluded from occurring.
Such PCR amplification can lead to significantly biased representation of divergent sequences, as some ces amplify more efficiently than others, which can lead to dramatic differences after the ntial amplification which occurs in PCR. The workflow described herein reduces this issue by having PCR products captured onto a bead. For example, if a cell’s VH and VL sequences are amplified very well or poorly, a r amount of product will be captured onto the bead. Thereby, there is a more even entation of antibody sequences in the final library, relative to methods that omit this step and perform linking by splicing by overlap extension PCR.
Definitions An “HC le region,” as that term is used herein, refers to a polypeptide comprising heavy chain CDRs 1, 2 and 3 and heavy chain FW regions 1, 2, 3, and 4.
An “LC variable region,” as that term is used herein, refers to a polypeptide comprising light chain CDRs 1, 2 and 3 and light chain FW regions 1, 2, 3, and 4.
A “heavy chain variable region sequence,” or “HCVRS,” as that term is used herein, refers to a polypeptide comprising sufficient sequence from heavy chain CDRs and sufficient sequence from heavy chain FW regions, to allow binding of antigen. In embodiments the HCVRS can assemble with a light chain variable region, and, e.g., bind antigen. In an embodiment, a HCVRS comprises sufficient sequence from heavy chain CDRs 1, 2, and 3,and sufficient sequence from heavy chain FW regions, e.g., heavy chain FW regions 1, 2, 3, and 4, to allow binding of antigen. In an ment, a HCVRS ses heavy chain CDRs 1, 2, and 3, and sufficient sequence from heavy chain FW regions, e.g., heavy chain FW s 1, 2, 3, and 4, to complex with a light chain variable region and to allow binding of antigen.
A “light chain variable region sequence,” or “LCVRS,” as that term is used herein, refers to a polypeptide comprising sufficient sequence from light chain CDRs and sufficient sequence from light chain FW regions, to allow binding of antigen. In embodiments the LCVRS can assemble with a heavy chain le region, and, e.g., bind antigen. In an embodiment, a LCVRS comprises sufficient sequence from light chain CDRs l, 2, and 3,and ient sequence from light chain FW regions, e.g., light chain FW regions 1, 2, 3, and 4, to allow binding of antigen. In an embodiment, a LCVRS comprises light chain CDRs 1, 2, and 3, and sufficient sequence from light chain FW regions, e.g., light chain FW regions 1, 2, 3, and 4, to x with a heavy chain variable region and to allow binding of antigen.
“Element” of an LC or HC variable region, as that term is used herein, refers to a sequence that encodes at least one amino acid. In an embodiment, an element comprises a CDR. In an embodiment an element comprises a FW region. In an embodiment, and element comprises a CDR and a FW region. In an embodiment an element comprises a HCVRS or a LCVRS. In an embodiment, the element comprises at least 10, 20, 30, 40, 50, 60, 70, 80, 90, or 100 amino acid residues.
A “micro-chamber,” as that term is used , refers to a compartment that is dimensioned, e.g., is sufficiently small, such that upon formation it ns a single cell, or the content from a single cell. In an ment, the micro-chamber has a volume of that is 10 to 10,000 times r of a cell that it contains. In an embodiment, the micro-chamber has a volume of 20 pL. In an embodiment, the micro-chamber has a maximum dimension of 100 nL. In an embodiment the micro- chamber comprises a droplet of liquid. In ment, the micro-chamber comprises a droplet of a first liquid disposed in an immiscible media, e.g., a gas or second . In an embodiment, the micro-chamber comprises a droplet of a first liquid, e. g., a lysis buffer or a PCR reaction , formed by dispersing the first liquid in an immiscible second liquid, e.g., a fluorinated oil. In an embodiment, the micro-chamber comprises a substrate and a nce other than the substrate, e.g., a solution. In an embodiment, the droplet comprises a substrate (e.g., a capture substrate, e.g., a bead) and a substance other than the substrate, e.g., a solution.
“Acquiring,” as that term is used herein, refers to possession of provision of an entity, e.g., a physical entity or data. Acquiring a physical entity includes making or manufacturing a physical entity tly acquiring) as well as receiving a physical entity from another party or source (indirectly acquiring). Acquiring a data or a value includes generating the data or value (directly acquiring) as well as ing the data or value from another party or source (indirectly acquiring).
“Matched,” as that term is used herein in connection with a heavy chain variable region and a light chain variable region, means they are from the same cell. With respect to an t of a light chain le region and an element of a heavy chain variable region it means that the light chain le region and the heavy chain variable region from which the elements are derived are from the same cell.
An “isolated reaction site,” as that term is used here, refers to a site, e.g., a location on a substrate, a micro chamber, or a well on a substrate, which allows for sufficient separation between a first loaded capture ate and a second loaded capture substrate, or generally, from HC or LC (or at chain or [3 chain, or 7 chain or 6 chain) encoding nucleic acid of another cell, such that the first loaded capture substrate is not contaminated with nucleic acid encoding a HC or LC (or 0t chain or [5 chain, or y chain or 5 chain) from another cell. In an embodiment an isolated reaction site provides sufficient separation between a first mRNA loaded capture substrate and a second mRNA loaded e substrate, or generally, from LC or HC encoding nucleic acid of another cell, that the first loaded mRNA capture substrate is not inated with nucleic acid, e. g., mRNA, encoding an HC or LC (or a chain or [3 chain, or 7 chain or 6 chain) from another cell. In an embodiment an ed reaction site provides sufficient separation n a first cDNA loaded capture substrate and a second cDNA loaded capture substrate, or generally, from HC or LC (or 0. chain or [3 chain, or y chain or 8 chain) encoding nucleic acid of another cell, that the first loaded cDNA capture substrate is not contaminated with nucleic acid, e.g., cDNA, encoding a HC or LC (or 0L chain or [3 chain, or 7 chain or 5 chain) from another cell. Separation can be provided, e.g., by sufficient distance n isolated reaction sites on a substrate; by configuring the isolated reaction sites such that they are not in fluid connection, or by formation of an immiscible barrier n a volume or chamber and the environment. In an embodiment, the isolated reaction site comprises a substrate and a substance other than the substrate, e.g., a solution.
“Complimentary,” as that term is used herein, refers to sequences which can form Watson- Crick pairing. When a first sequence is complementary with a second ce it can be mentary to the entire second sequence or to less than all of the second sequence.
A “display entity,” as that term is used herein, refers to an entity, e. g., a phage or cell, e. g., a yeast cell, which es a gene that encodes a polypeptide.
An “AC variable region,” as that term is used herein, refers to a polypeptide comprising TCR 0t chain CDRs l, 2 and 3 and 0t chain FW regions 1, 2, 3, and 4.
A “BC le region,” as that term is used herein, refers to a ptide comprising [3 chain CDRs 1, 2 and 3 and [3 chain FW regions 1, 2, 3, and 4.
A “GC variable region,” as that term is used , refers to a polypeptide comprising TCR V chain CDRs 1, 2 and 3 and 7 chain FW regions 1, 2, 3, and 4.
A “DC variable region,” as that term is used herein, refers to a polypeptide comprising 5 chain CDRs l, 2 and 3 and 5 chain FW regions 1, 2, 3, and 4.
An “(1 chain variable region sequence,” or “ACVRS,” as that term is used herein, refers to a polypeptide comprising sufficient sequence from 0t chain CDRs and sufficient ce from 0. chain FW regions, to allow g of antigen. In ments the ACVRS can assemble with a [3 chain variable region, and, e.g., bind antigen. In an embodiment, a ACVRS comprises sufficient sequence from or chain CDRs 1, 2, and 3, and sufficient sequence from or chain FW s, e.g., or chain FW regions 1, 2, 3, and 4, to allow binding of antigen. In an embodiment, an ACVRS ses 0. chain CDRs 1, 2, and 3, and sufficient sequence from or chain FW regions, e.g., 0. chain FW regions 1, 2, 3, and 4, to complex with a [3 chain variable region and to allow binding of antigen.
A “[3 chain variable region sequence,” or “BCVRS,” as that term is used herein, refers to a polypeptide comprising sufficient sequence from [3 chain CDRs and sufficient sequence from [3 chain FW regions, to allow binding of n. In embodiments the BCVRS can assemble with an 0. chain variable region, and, e.g., bind antigen. In an embodiment, a BCVRS comprises sufficient sequence from [3 chain CDRs 1, 2, and 3, and sufficient sequence from [5 chain FW regions, e.g., [3 chain FW regions 1, 2, 3, and 4, to allow binding of antigen. In an embodiment, a BCVRS comprises [3 chain CDRs 1, 2, and 3, and sufficient sequence from [3 chain FW s, e.g., [3 chain FW regions 1, 2, 3, and 4, to complex with an a chain variable region and to allow binding of antigen.
A “7 chain variable region sequence,” or “GCVRS,” as that term is used herein, refers to a polypeptide comprising sufficient sequence from 7 chain CDRs and sufficient sequence from 7 chain FW regions, to allow binding of antigen. In ments the GCVRS can assemble with a 5 chain variable region, and, e.g., bind antigen. In an embodiment, a GCVRS comprises sufficient sequence from 7 chain CDRs 1, 2, and 3, and sufficient sequence from y chain FW regions, e.g., y chain FW regions 1, 2, 3, and 4, to allow binding of antigen. In an embodiment, a GCVRS comprises 7 chain CDRs l, 2, and 3, and sufficient sequence from y chain FW regions, e.g., 7 chain FW regions 1, 2, 3, and 4, to complex with a 5 chain variable region and to allow binding of antigen.
A “5 chain variable region ce,” or “DCVRS,” as that term is used herein, refers to a polypeptide comprising sufficient sequence from 5 chain CDRs and sufficient sequence from 5 chain FW s, to allow binding of antigen. In embodiments the DCVRS can assemble with a y chain variable , and, e.g., bind antigen. In an embodiment, a DCVRS comprises sufficient sequence from 5 chain CDRs 1, 2, and 3,and sufficient sequence from 5 chain FW regions, e.g., 5 chain FW regions 1, 2, 3, and 4, to allow binding of antigen. In an embodiment, a DCVRS comprises 5 chain CDRs 1, 2, and 3, and sufficient sequence from 5 chain FW regions, 6g, 5 chain FW s 1, 2, 3, and 4, to complex with an or chain variable region and to allow binding of antigen.
“Element” of an 0. chain or [3 chain le region, as that term is used herein, refers to a sequence that encodes at least one amino acid. In an embodiment, an element comprises a CDR. In an embodiment an element ses a FW region. In an embodiment, and element comprises a CDR and a FW region. In an embodiment an element comprises an ACVRS or a BCVRS. In an embodiment, the t comprises at least 10, 20, 30, 40, 50, 60, 70, 80, 90, or 100 amino acid “Element” of a 7 chain or 5 chain variable region, as that term is used herein, refers to a sequence that encodes at least one amino acid. In an embodiment, an element comprises a CDR. In an embodiment an element comprises a FW region. In an embodiment, and element comprises a CDR and a FW region. In an embodiment an element comprises a GCVRS or a DCVRS. In an embodiment, the element ses at least 10, 20, 30, 40, 50, 60, 70, 80, 90, or 100 amino acid residues.
“Matched,” as that term is used herein in connection with an or chain le region and a [5 chain variable region, means they are from the same cell. With respect to an element of an 0. chain variable region and an element of a [3 chain variable region it means that the or chain variable region and the [3 chain variable region from which the elements are derived are from the same cell.
“Matched,” as that term is used herein in connection with a 7 chain variable region and a 5 chain variable region, means they are from the same cell. With respect to an t of a y chain variable region and an element of a 5 chain variable region it means that the 7 chain variable region and the 7 chain variable region from which the elements are derived are from the same cell.
As used herein, the articles “a” and “an” refer to one or to more than one (e.g., to at least one) of the grammatical object of the article.
The term “or” is used herein to mean, and is used interchangeably with, the term “and/or”, unless context clearly indicates otherwise.
“About” and “approximately” shall generally mean an acceptable degree of error for the quantity measured given the nature or ion of the measurements. Exemplary degrees of error are within 20 percent (%), typically, within 10%, and more typically, within 5% of a given value or range of values.
The compositions and methods disclosed herein ass polypeptides and nucleic acids having the sequences specified, or sequences substantially identical or similar thereto, e.g., sequences at least 85%, 90%, 95% identical or higher to the sequence specified.
In the context of an amino acid sequence, the term antially identical” is used herein to refer to a first amino acid that contains a sufficient or m number of amino acid residues that are i) identical to, or ii) conservative tutions of aligned amino acid residues in a second amino acid sequence such that the first and second amino acid sequences can have a common structural domain and/or common functional activity. For example, amino acid sequences that contain a common structural domain having at least about 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identity to a reference sequence, e.g., a sequence provided herein.
In the context of nucleotide sequence, the term “substantially identical” is used herein to refer to a first nucleic acid ce that contains a sufficient or m number of nucleotides that are identical to aligned nucleotides in a second nucleic acid ce such that the first and second nucleotide sequences encode a polypeptide having common functional activity, or encode a common structural polypeptide domain or a common functional polypeptide activity. For example, nucleotide sequences having at least about 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identity to a reference ce, e.g., a sequence provided herein.
The term “functional variant” refers polypeptides that have a substantially identical amino acid sequence to the naturally-occurring sequence, or are encoded by a substantially identical nucleotide sequence, and are capable of having one or more activities of the naturally-occurring sequence.
Calculations of homology or sequence identity between sequences (the terms are used interchangeably herein) are performed as follows.
To determine the percent identity of two amino acid sequences, or of two nucleic acid sequences, the sequences are aligned for optimal comparison purposes (e.g., gaps can be uced in one or both of a first and a second amino acid or nucleic acid sequence for optimal alignment and non-homologous sequences can be disregarded for ison purposes). In a typical embodiment, the length of a reference sequence aligned for comparison purposes is at least 30%, e.g., at least 40%, 50%, 60%, e.g., at least 70%, 80%, 90%, 100% of the length of the reference sequence. The amino acid residues or nucleotides at ponding amino acid positions or nucleotide positions are then compared. When a position in the first ce is occupied by the same amino acid e or nucleotide as the corresponding position in the second sequence, then the molecules are identical at that on.
The percent identity between the two sequences is a function of the number of identical positions shared by the sequences, taking into t the number of gaps, and the length of each gap, which need to be introduced for optimal ent of the two sequences.
The ison of sequences and determination of percent identity n two sequences can be accomplished using a mathematical algorithm. In some embodiments, the percent identity between two amino acid sequences is determined using the Needleman and Wunsch ((1970) J. M01.
Biol. -453) algorithm which has been incorporated into the GAP program in the GCG software package (available at www.gcg.com), using either a Blossum 62 matrix or a PAM250 matrix, and a gap weight of 16, 14, 12, 10, 8, 6, or 4 and a length weight of l, 2, 3, 4, 5, or 6. In certain embodiments, the percent identity between two nucleotide sequences is determined using the GAP program in the GCG software package (available at www.gcg.com), using a NWSgapdna.CMP matrix and a gap weight of 40, 50, 60, 70, or 80 and a length weight of l, 2, 3, 4, 5, or 6. One suitable set of parameters (and the one that should be used unless otherwise specified) are a Blossum 62 scoring matrix with a gap penalty of 12, a gap extend penalty of 4, and a frameshift gap penalty of 5.
The percent identity between two amino acid or nucleotide sequences can be determined using the algorithm of E. Meyers and W. Miller ((1989) CABIOS, 4: 11-17) which has been incorporated into the ALIGN program (version 2.0), using a PAM120 weight residue table, a gap length y of 12 and a gap penalty of 4.
The nucleic acid and n ces described herein can be used as a “query sequence” to perform a search against public ses to, for example, identify other family members or related sequences. Such searches can be med using the NBLAST and XBLAST programs on 2.0) of Altschul, et al. (1990) J. Mol. Biol. 215:403-10. BLAST nucleotide searches can be performed with the NBLAST m, score = 100, wordlength = 12 to obtain nucleotide sequences homologous to a nucleic acid as described . BLAST protein searches can be med with the XBLAST program, score = 50, wordlength = 3 to obtain amino acid sequences homologous to protein molecules described herein. To obtain gapped alignments for comparison purposes, Gapped BLAST can be utilized as described in Altschul et al., (1997) Nucleic Acids Res. 25 :3389-3402. When utilizing BLAST and gapped BLAST programs, the default parameters of the respective programs (e.g., XBLAST and NBLAST) can be used. See www.ncbi.nlm.nih.gov.
As used herein, the term “hybridizes under low stringency, medium stringency, high ency, or very high stringency conditions” describes conditions for hybridization and washing. ce for ming hybridization reactions can be found in Current Protocols in Molecular Biology, John Wiley & Sons, NY. (1989), 63.1-63.6, which is incorporated by reference. Aqueous and nonaqueous methods are described in that nce and either can be used. Specific hybridization conditions referred to herein are as s: 1) low stringency hybridization conditions in 6X sodium chloride/sodium citrate (SSC) at about 45°C, followed by two washes in 0.2X SSC, 0.1% SDS at least at 50°C (the temperature of the washes can be increased to 55°C for low stringency conditions); 2) medium stringency hybridization conditions in 6X SSC at about 45°C, followed by one or more washes in 0.2X SSC, 0.1% SDS at 60°C; 3) high stringency hybridization conditions in 6X SSC at about 45°C, followed by one or more washes in 0.2X SSC, 0.1% SDS at 65°C; and ably 4) very high stringency hybridization conditions are 0.5M sodium phosphate, 7% SDS at 65°C, followed by one or more washes at 0.2X SSC, 1% SDS at 65°C. Very high stringency conditions 4) are suitable conditions and the ones that should be used unless otherwise specified.
It is understood that the molecules described herein may have additional conservative or non- essential amino acid substitutions, which do not have a substantial effect on their functions.
The term “amino acid” is intended to embrace all molecules, whether natural or synthetic, which include both an amino functionality and an acid functionality and capable of being ed in a polymer of lly-occurring amino acids. Exemplary amino acids include naturally-occurring amino acids; analogs, tives and congeners thereof; amino acid analogs having variant side chains; and all stereoisomers of any of any of the foregoing. As used herein the term “amino acid” includes both the D- or L- optical isomers and peptidomimetics.
A “conservative amino acid substitution” is one in which the amino acid residue is replaced with an amino acid residue having a similar side chain. es of amino acid residues having similar side chains have been defined in the art. These families include amino acids with basic side chains (e.g., lysine, arginine, ine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (6.3., glycine, asparagine, ine, serine, threonine, tyrosine, cysteine), nonpolar side chains (e. g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), beta-branched side chains (e.g., threonine, valine, cine) and aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan, histidine).
The terms eptide,93 upeptide” and “protein” (if single chain) are used interchangeably herein to refer to polymers of amino acids of any length. The polymer may be linear or branched, it may comprise modified amino acids, and it may be interrupted by ino acids. The terms also encompass an amino acid polymer that has been modified; for example, disulfide bond formation, glycosylation, lipidation, acetylation, orylation, or any other lation, such as conjugation with a labeling ent. The polypeptide can be ed from natural sources, can be a produced by inant techniques from a eukaryotic or prokaryotic host, or can be a product of synthetic procedures. In an embodiment, the polypeptide is an antibody molecule. In another embodiment, the polypeptide is a TCR molecule, e.g., soluble TCR molecule.
The terms “nucleic acid,” “nucleic acid sequence,a, unucleotide sequence,” or “polynucleotide sequence,” and “polynucleotide” are used interchangeably. They refer to a polymeric form of nucleotides of any length, either deoxyribonucleotides or ribonucleotides, or analogs thereof. The polynucleotide may be either single-stranded or double-stranded, and if single-stranded may be the coding strand or non-coding (antisense) . A polynucleotide may comprise d nucleotides, such as methylated nucleotides and nucleotide analogs. The sequence of nucleotides may be interrupted by non-nucleotide components. A polynucleotide may be further modified after polymerization, such as by conjugation with a labeling component. The nucleic acid may be a recombinant polynucleotide, or a polynucleotide of genomic, cDNA, semisynthetic, or synthetic origin which either does not occur in nature or is linked to another polynucleotide in a non-natural arrangement.
The term “isolated,” as used herein, refers to material that is removed from its original or native nment (e.g., the natural environment if it is lly occurring). For example, a lly-occurring polynucleotide or polypeptide present in a living animal is not isolated, but the same polynucleotide or polypeptide, ted by human intervention from some or all of the co- existing materials in the l system, is isolated. Such polynucleotides could be part of a vector andior such polynucleotides or polypeptides could be part of a composition, and still be isolated in that such vector or composition is not part of the environment in which it is found in .
As used herein, the term “treat,” e.g., a disorder described herein, means that a subject (e.g., a human) who has a disorder, e.g., a disorder described herein, and/or experiences a symptom of a disorder, e.g., a disorder described herein, will, in an embodiment, suffer less a severe symptom and/or recover faster when an antibody molecule is administered than if the antibody molecule were never stered. Treatment can, e.g., partially or completely, alleviate, rate, relieve, inhibit, or reduce the severity of, and/or reduce incidence, and optionally, delay onset of, one or more manifestations of the effects or symptoms, features, and/or causes of the disorder. In an embodiment, treatment is of a subject who does not exhibit certain signs of the disorder, and/or of a subject who exhibits only early signs of the disorder. In an ment, treatment is of a subject who exhibits one or more ished signs of a disorder. In an embodiment, treatment is of a subject diagnosed as ing from a disorder.
As used herein, the term “prevent,” a disorder, means that a subject (e.g., a human) is less likely to have the disorder, if the subject receives a polypeptide (e.g., antibody molecule).
Various aspects of the compositions and methods herein are described in further detail below.
Additional definitions are set out throughout the specification.
Libraries of Binding Polypeptides Disclosed herein are libraries (e.g., y libraries) of binding polypeptides, e. g., antibody les or T cell receptor molecules, and methods of making libraries of binding polypeptides, e.g., antibody molecules or T cell receptor molecules.
In an embodiment, a method described herein links two DNA fragments, such as sequences encoding an antibody heavy chain variable region (or a portion thereof) and an dy light chain variable region (or a portion thereof), a TCR or chain (or a portion thereof) and a TCR [3 chain (or a portion thereof), or a TCR 7 chain (or a portion thereof) and a TCR 5 chain (or a n thereof), using a ligase-mediated approach.
For example, antibodies are composed of two types of ptide chains, light chain and heavy chain, each of which are ated from separate mRNA molecules. In order to copy a functional unit of a ular antibody (or B cell receptor) from a B cell, knowledge of the particular heavy chain and its cognate light chain must be ined. This is lly med using methods in which individual clones are in wells of microwell plates, which keeps clones segregated and the result heavy and light chain sequences thus are known to be paired. Such cloning processes scale well to B cell numbers compatible with 96- or 384-well plates. However, B cell repertoires in humans and animals can range from about 106-1011 B cells, many of which are different clones (La, different BCRs or antibodies). Thus, there is a need to be able to, in an efficient manner, make copies of millions to billions of B cells which (1) retains native pairing of chains and (2) allows for functional interrogation of such a large number of unique clones. Such a method can facilitate making a renewable copy of an antibody oire which can be functionally interrogated by a variety of methods.
In an embodiment, a method described herein uses one or more (e.g., two, three, or all) of the following: (1) miniaturized compartmentalization of individual cells (e.g., B cells or T cells) in droplets (pL to nL volume drops), (2) lysing and PCR amplifying two chains (e.g., antibody VH and VL, TCR 0t and [3 chains, or TCR V and 5 chains), (3) specifically linking the two , such that native chain pairing is retained and that a thermostable ligase catalyzes the linking, and (4) amplifying the linked DNA in a manner that allows for high throughput phenotypic ogation of clones by a surface display technology, such as yeast or phage display.
The s bed herein can result in a nucleic acid sequence, when expressed, encodes a functional polypeptide, e.g., a functional antigen binding polypeptide. For e, the HC element and the LC element (or the AC element and the BC element, or the GC element and the DC element) are not configured in a head-to-head or tail-to-tail ation. In an embodiment, the HC element and the LC element (or the AC element and the BC element, or the GC t and the DC element) are configured in a o-tail orientation. For example, the C-terminus of the LC element (or LCVRS) is linked, directly or indirectly, with the N-terminus of the HC element (or HCVRS), or the C- terminus of the HC element (or HCVRS) is linked, directly or indirectly, with the C-terminus of the LC element (or LCVRS).
Exemplary Workflow Cells (e.g., immune cells, e.g., B cells or T cells) are encapsulated dually into drops. In the drops, the cells are lysed and mRNA is captured onto beads, which contain oligonucleotides to hybridize to mRNA. The beads tate maintaining native pairing information (e.g., the native pairing n two chains, e.g., a heavy chain and a light chain in a single B cell; an 0t chain and a [3 chain in a single T cell; or a 7 chain and a 6 chain in a single T cell). Next, the mRNA is reversed transcribed to cDNA by a reverse transcriptase (RT). The e transcription can be performed within the lysis drops, outside of drops, or in the subsequent drop (‘PCR’ drop). Beads having captured mRNA or cDNA are recovered from the initial drops. The beads are then ulated into new drops, wherein the nucleic acids are amplified, either by RT-PCR (when mRNA is template) or PCR (when cDNA is template). The cDNAs encoding the two chains are amplified in drops. The ied products are captured back onto beads by specific complementary nucleic acid hybridization. The beads having captured products are recovered from drops and subsequently encapsulated into new drops. The amplified product encoding one chain (e.g., VH) is linked with the amplification product encoding the other chain (e. g., VL) in drops using a thermostable ligase. In an approach (“linking cohesive products”), cohesive (or “sticky-end”) PCR products are generated, and covalent ligation of hybridized cohesive PCR products are performed by a thermostable ligase. In another ch (“ligase cycling reaction”), no cohesive PCR products are produced. Rather, in drops, DNA is linked together through use of a stable ligase and a splint (or bridging) oligonucleotide. While not wishing to be bound by theory, it is believed that in an embodiment, the methods described herein reduce or preclude the possibility of unintended fusing caused by overlap extension PCR methods (Turchaninova et al. Eur J Immunol. 2013; 43(9): 2507-2515). The ligated ts, representing natively paired chains, are further amplified to generate sufficient al to create a display library, such as in yeast or phage. The amplified product, encoding natively paired chains (e.g., antibody heavy chain and light chain, TCR 0L chain and [3 chain, or TCR 7 chain and 5 chain) in a format such as an scFv, scFab, Fab, or full-length IgG, are introduced to an appropriate expression or display vehicle, such as yeast or phage display. The ucted y, e. g., having >104 and up to 109 or larger members, can be rapidly interrogated for desired binding and/or other phenotypic properties, using established methods.
Generation of Cohesive PCR Products That Are Suitable Substratesfor Ligase In an embodiment, amplification (e.g., PCR) products with cohesive ends that are suitable substrates for ligase are generated. Without wishing to be bound by theory, it is believed that in an embodiment, DNA polymerase extension can be prematurely terminated at a defined location, e.g., through use of a chemically d (e.g., lesioned) nucleotide or base, or other alterations to the primer used for amplification. These chemically modified nucleotides or bases (or other primer alterations) are subsequently incorporated into one strand of the amplification t. As the DNA polymerase reads h the template strand which contains the d nucleotide, it prematurely stops extension at (or near) the modified nucleotide, as it is not able to read through. This early polymerase termination due to the modification can lead to production of an ication product with a cohesive end. The amplification product can hybridize (or anneal) ntly with another ication product having a complementary ve end, which can be produced in an analogous matter. For example, a PCR product encoding one chain (6.3., VH) and a PCR product encoding another chain (e.g., VL), each having a complementary cohesive end, can hybridize (or anneal) to each other with high efficiency. Next, a thermostable ligase, t in the droplet with the DNA polymerase (e. g., throughout thermocycling), catalyzes ligation ent linkage) of the hybridized (or annealed) DNA molecules.
In an embodiment, the native paring ation is maintained during amplification and ligation. In an embodiment, both amplification and ligation occur in the same drop, e.g., without breaking the drop. In an embodiment, the ligase retains at least 50%, e.g., at least 60%, 70%, 80%, 85%, 90%, 95%, or 99% activity, at 95°C or more (e.g., 96°C or more, 97°C or more, or 98°C or more), during one or more (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, or more) thermocycles. In an embodiment, the ligase retains at least 50%, e.g., at least 60%, 70%, 80%, 85%, 90%, 95%, or 99% activity, at 95°C or more (e.g., 96°C or more, 97°C or more, or 98°C or more), for at least 5 s (e. g., at least 10 minutes, 15 minutes, 20 minutes, 25 s, 30 s, 45 minutes, or 60 minutes).
In an ment, the ligase retains at least 50%, e.g., at least 60%, 70%, 80%, 85%, 90%, 95%, or 99% activity, at 95°C or more (e.g., 96°C or more, 97°C or more, or 98°C or more), in a buffer ion that allows for DNA polymerase activity.
In an embodiment, the modification inhibits or blocks DNA polymerase activity and remains a substrate for ligation. In an embodiment, the modification does not inhibit or prevent g of the ication product to a ligase. In an embodiment, the modification does not inhibit or prevent formation of a phosphodiester bond. In an embodiment, the modification does not comprise a large bulky chemical group. Exemplary modifications include, but are not limited to, a ribose 2’-C (2nd carbon) modification (e.g., OH (516., a ribonucleotide, not a deoxyribonucleotide), O-methyl (O-CH3), or amine (NH2)); a ribose 4’-C (4th carbon) modification; a base modification (e.g., a non-native base, a uracil, or others); an abasic site (e.g., an AP site or apyrimidine/apurine); or a staggered primer (e.g., different length ng). In an embodiment, the modification comprises a uracil and a DNA polymerase that is inhibited by uracil (e.g., an archaeal DNA polymerase) is used for amplification.
Exemplary steps for performing a cohesive-end PCR-ligation ment in drops are illustrated below.
Cell Encapsulation Cells can be encapsulated individually into droplets. In an embodiment, the cell is an immune cell. In an embodiment, the cell is a B cell. In an embodiment, the cell is a T cell. In an embodiment, the cell is an antibody-producing cell. In an embodiment, the cell is an isolated cell or purified cell.
In an embodiment, the cell is obtained from a subject, e.g., a human, mouse, rabbit, rat, goat, sheep, or chicken.
In an embodiment, the volume of the droplet is from 10 pL to 100 nL, e.g., from 10 pL to 100 pL, from 10 pL to 1000 pL, from 10 pL to 10 nL, from 10 nL to 100 nL, from 1000 pL to 100 nL, from 100 pL to 100 nL, from 100 pL to 10 nL, from 100 pL to 1000 pL, from 1000 pL to 10 nL, or from 100 pL to 1000 pL. In an embodiment, the volume of the droplet is from 100 pL to 1000 pL.
In an embodiment, the droplet is a water-in-oil droplet. In an embodiment, the droplet is present in a r (e.g., oil) phase, e.g., a carrier phase comprising 3MTM HFE-7500 with about 1% fiuorosurfactant (RAN Biotechnologies).
The droplets can be formed, e. g., using a microfluidic chip (e.g., 2R 100 pm from te) with the flow of fluid phase controlled by a syringe or pressure pump. In an embodiment, the aqueous phase of the droplet comprises a buffer, a reagent that aids cell lysis, and a bead. In an embodiment, the buffer ses Tris at pH 7.5. In an embodiment, the reagent that aids cell lysis comprises a detergent. Exemplary detergents that can be used to aid cell lysis include, but are not limited to, Tween-20, Triton X, IGEPAL, or sodium lauroyl sarcosinate (Sarkosyl). In an embodiment, the bead is a magnetic bead. In an ment, the bead comprises, is coupled to, an oligonucleotide (e. g., a primer), e. g., to anneal to an mRNA (e. g., an mRNA encoding a heavy chain or a light chain).
In an embodiment, the droplet contains no more than one cell after encapsulation. In an embodiment, the droplet contains a ity of beads. In an embodiment, a plurality of beads are obtained, and at least 80%, e. g., at least 85%, 90%, 95 %, 98%, 99%, or 100%, of the plurality contains no more than one cell per droplet. In an embodiment, a plurality of beads are obtained, and at least 80%, e. g., at least 85%, 90%, 95%, 98%, 99%, or 100%, of the plurality contains at least one bead per droplet. Typically, the occupancy of drops is no more than one cell per droplet, and at least one bead per droplet.
Cell Lysis After encapsulation, the droplets can be ted to facilitate cell lysis. In an embodiment, an emulsion (e. g., containing ce different solution phases) is heated, e. g., to reduce mRNA secondary structures so that it can be more efficiently captured onto the bead and/or to improved lysis efficiency in the presence of a detergent (e. g., Tween20). In an embodiment, the emulsion is incubated at a temperature n 40°C and 80°C, e. g., between 40°C and 60°C, 50°C and 70°C, or 60°C and 80°C, e. g., at 40°C, 50°C, 60°C, 70°C, or 80°C. In an embodiment, the emulsion is incubated for 5 to 60 minutes, e.g., 10 to 45 minutes, 15 to 30 minutes, 5 to 30 minutes, or 30 to 50 minutes. In an ment, the cell is lysed by heat. In an embodiment, the cell is lysed by an enzyme. Typically, after the cell is lysed, mRNA is released and is captured on a bead by annealing to the oligonucleotides on the bead.
Bead ry Emulsions (e.g., containing coalesce different solution phases) can be broken using a drop ilizing reagent, e.g., perfluorooctanol (PFO). In an embodiment, the bead-containing aqueous phase is recovered. In an embodiment, the bead is a magnetic bead, and is isolated using magnet. In an ment, the bead is washed and resuspended in a buffer (e. g., Tris, pH 7.5).
Reverse Transcription Reverse transcription can be performed using standard methods. In an embodiment, the reverse transcription is performed in a ulsion reaction. In an embodiment, the reverse transcription is performed in an emulsion reaction. In an ment, the bead with captured mRNA is resuspended in a buffer-enzyme mix (e.g., Superscript II RT) and incubated at 35°C to 45°C (e. g., at 40°C) for 10 to 60 minutes (e.g., 15 minutes) to facilitate reverse ription. In an ment, the oligonucleotide coupled to the bead is used as a primer for the synthesis of the first strand cDNA.
In an embodiment, the bead is washed with a buffer (e.g., Tris, pH 7.5) after reverse transcription.
Bead Encapsulation Beads can be ulated individually into droplets. In an embodiment, the volume of the droplet is from 5 pL to 500 pL, e. g., from 5 pL to 400 pL, from 5 pL to 300 pL, from 5 pL to 200 pL, from 5 pL to 100 pL, from 5 pL to 50 pL, from 5 pL to 25 pL, from 400 pL to 500 pL, from 300 pL to 500 pL, from 200 pL to 500 pL, from 100 pL to 500 pL, from 50 pL to 500 pL, from 25 pL to 500 pL, from 10 pL to 500 pL, from 10 pL to 400 pL, from 25 pL to 300 pL, from 50 pL to 200 pL, or from pL to 50 pL. In an embodiment, the volume of the droplet is from 10 pL to 50 pL.
In an embodiment, the droplet is a water-in-oil droplet. In an ment, the droplet is present in a carrier (e.g., oil) phase, e.g., a carrier phase comprising 3MTM HFE-7500 with about 1% urfactant (RAN Biotechnologies).
In an embodiment, the droplet contains one bead after encapsulation. In an embodiment, a plurality of beads are obtained, and at least 80%, e.g., at least 85%, 90%, 95%, 98%, 99%, or 100%, of the plurality contains no more than one bead per droplet.
PCR-Ligarion Reaction In an ment, a PCR-ligation reaction is performed. In an embodiment, the PCR- ligation reaction generates a ligated product (e.g., a -stranded DNA) that comprises a nucleotide sequence that encodes an antibody heavy chain variable region (or a n thereof) and an antibody light chain variable region (or a portion thereof). In an embodiment, the PCR-ligation reaction generates a d product (e.g., a double-stranded DNA) that comprises a nucleotide sequence that encodes a TCR (1 Chain (or a portion thereof) and a TCR [3 chain (or a portion thereof).
In an embodiment, the PCR-ligation reaction generates a ligated product (e.g., a double-stranded DNA) that comprises a nucleotide sequence that encodes a TCR 7 chain (or a portion thereof) and a TCR 5 chain (or a portion thereof). In an embodiment, the PCR-ligation reaction is performed in a droplet comprising a bead that is d with cDNA, a DNA polymerase, oligonucleotides (e.g., for amplification of the cDNA), a ligase (e.g., a thermostable ), and a .
Exemplary DNA polymerases that can be used in the reaction include, but are not d to, n® High-Fidelity DNA Polymerase (NEB), Q5® High-Fidelity DNA Polymerase (NEB), Pfu DNA polymerase, KAPA DNA polymerase, Vent® DNA polymerase, or Taq DNA polymerase.
In an ment, the ligated product contains an scFv, a Fab, or scFab cassette. In an embodiment, the te (e.g., scFv cassette) is constructed as VL-Linker-VH. Without wishing to be bound by theory, it is believed that in an embodiment, the order can be switched to VH-Linker-VL, with no significant impact on expression or function. In an embodiment, the cassette (e.g., scFv) cassette is constructed as VH-Linker-VL. In an embodiment, the cassette comprises a constant region sequence (e.g., a CH1 domain and/or a CL domain), e.g., VH-CHl coupled with VL-CL.
Similarly, the ligated t can contain a cassette constructed as at chain-Linker-B chain or [3 chain-Linker-a chain, or y chain-Linker-5 chain or 5 chain-Linker-y chain.
In an embodiment, the reverse primer for the VL sequence contains one, two, or all of the following: (a) an ng sequence encoding a linker sequence (b) at least one modified nucleotide (e.g., 3 consecutive nucleotides with 2’-O-methy1 modification), e.g., in the overhang; or (c) a 5’- phosphate. In an embodiment, the forward primer for the VH sequence contains one, two, or all of the following: (a) an ng sequence encoding a linker sequence (b) at least one modified tide (e.g., 3 consecutive nucleotides with 2’-O-methyl modification), e.g., in the overhang; or (c) a sphate.
In an embodiment, the reverse primer for the VH ce contains one, two, or all of the following: (a) an overhang ce encoding a linker sequence (b) at least one modified nucleotide (e. g., 3 consecutive nucleotides with 2’-O-methyl modification), e.g., in the overhang; or (c) a 5’- phosphate. In an embodiment, the forward primer for the VL sequence contains one, two, or all of the following: (a) an overhang sequence encoding a linker sequence (b) at least one modified nucleotide (6.3., 3 consecutive nucleotides with 2’-O-methyl modification), e.g., in the overhang; or (c) a 5’- phosphate.
Similarly, in an ment, the reverse primer for the OL chain (or 7 chain) sequence ns one, two, or all of the following: (a) an overhang sequence encoding a linker ce (b) at least one modified nucleotide (e.g., 3 consecutive nucleotides with 2’-O-methyl modification), e.g., in the overhang; or (c) a 5’-phosphate. In an embodiment, the forward primer for the B chain (or 6 chain) sequence contains one, two, or all of the ing: (a) an overhang ce ng a linker sequence (b) at least one modified nucleotide (e. g., 3 consecutive nucleotides with 2’-O-methyl modification), e.g., in the overhang; or (c) a sphate.
Similarly, in an embodiment, the e primer for the [3 chain (or 5 chain) sequence contains one, two, or all of the following: (a) an overhang sequence encoding a linker sequence (b) at least one modified nucleotide (e.g., 3 consecutive nucleotides with 2’-O-methyl modification), e.g., in the overhang; or (c) a 5’-phosphate. In an embodiment, the forward primer for the 0: chain (or 7 chain) sequence contains one, two, or all of the following: (a) an overhang sequence encoding a linker sequence (b) at least one modified nucleotide (e.g., 3 consecutive tides with 2’-O-methyl modification), e.g., in the ng; or (c) a 5’-phosphate.
Exemplary ligases (e.g., thermostable ligases) that can be used in the reaction include, but are not limited to, Taq DNA ligase, Pfu DNA ligase, Ampligase® thermostable DNA ligase, Tsc DNA ligase, Rma DNA ligase, Tfi DNA ligase, or Tth DNA ligase.
In an embodiment, the buffer supports both DNA polymerase and ligase enzymatic activities.
In an embodiment, the thermocycling is performed with emulsion (e.g., in a PCR tube). In an embodiment, the thermocycling is performed using the following conditions: initial denaturation at 95-98°C for 30 seconds to 2 minutes; 10-30 cycles of: ration at 95-98°C for 10-30 seconds, primer annealing at 50-60°C for 10-30 seconds, polymerase ion at 72°C for 30 seconds, and cohesive product annealing and ligation at 45-55°C for 3 minutes. The reaction can be hold at 4°C.
Recovery ofAqueous Portion Emulsions (e.g., containing coalesce different solution phases) can be broken using a drop destabilizing reagent, e. g., perfluorooctanol (PFO). In an embodiment, the aqueous portion (e. g., containing linked product, and optionally, nked product) is recovered. In an embodiment, the bead is discarded.
Purification ofLinked t Linked product (e.g., enting natively linked VL—linker-VH) can be purified from non- linked products (e. g., non-linked VH and VL). The ligated products are separated from non-ligated products by size separation. For example, denaturing PAGE crylamide gel ophoresis) or denaturing HPLC-SEC can be used. The linked product (~800-900 bp) is isolated from non-linked product (~350-500 bp). For denaturing PAGE purification, the ligated band is cut out from the gel and an elecro-elution is med to extract DNA from gel slice (Bio-Rad Electro-Elutor).
Amplification ofPurified Linked Product The purified linked product can be amplified, e. g., by PCR. For example, the purified linked product is amplified by PCR using a DNA polymerase (e.g., Taq polymerase) under conditions that can moderately read through DNA containing modified nucleotides.
The final PCR product can be introduced to yeast using standard methods (e.g., electroporation with expression vector) to create a ly paired library d from biological SOUICCS.
Ligase Cycling In an embodiment, the different chains (e. g., VH and VL) are not amplified in a manner which incorporates DNA sequence common to both chains, which would facilitate annealing of sticky-end ts directly to each other. In an embodiment, a bridging (or ) oligonucleotide is included after the amplification of cDNA but in the presence of a thermostable ligase. The bridging oligonucleotide can facilitate bringing the two chains immediately adjacent to each other such that they become a substrate of . Ligase, in turn, catalyzes a covalent bond formation between chains of DNA. Since this mechanism does not lead to incorporation of sequence common to both chains in each chain (e.g., overhang DNA with common sequence in both VH and VL), there is no opportunity for splicing by p ion PCR.
The steps of this approach are generally the same as above except beginning at the emulsion PCR step. PCR amplification of cDNA can be performed in drops. s can add overhang sequences, but there is generally no common sequence to both chains (e.g., VL and VH) that is added (unlike the above strategy). The bead in the drop, through its ated oligonucleotides, becomes filled or saturated with dsDNA ts of the two chains (e.g., VH and VL), each with specific overhang sequence. Drops are broken, and any PCR product not linked or annealed to beads is washed away. Beads containing dsDNA of two chains (e.g., VH and VL) are ulated into new drops in the presence of a thermostable ligase and a splint oligonucleotide. In this emulsion format, cycling is performed, which facilitates formation of the 3-DNA piece complex. This complex is a substrate for ligase, which catalyzes covalent bond formation, linking the two . In an embodiment, thermocycling aids sion of all ‘top strand’ DNA to linked product, until one substrate becomes limiting. In another embodiment, both s become ligated. For example, once the ‘top strand’ is d, it can serve as the ‘splint’ for the opposing strand, which ligase will recognize as a substrate. Without wishing to be bound by theory, it is believed that, this facet specifically makes the reaction nt, that is, initial ligated product can serve as more templates (splints) to te even more additional d product. Drops are , and the ligated products are amplified by rd PCR means. ary steps for performing a ligase cycling experiment are illustrated below.
Cell Encapsulation Cells can be encapsulated individually into droplets. In an embodiment, the cell is an immune cell. In an embodiment, the cell is a B cell. In an embodiment, the cell is a T cell. In an embodiment, the cell is an antibody-producing cell. In an embodiment, the cell is an isolated cell or purified cell.
In an embodiment, the cell is obtained from a subject, e.g., a human, mouse, rabbit, rat, goat, sheep, or chicken.
In an embodiment, the volume of the droplet is from 10 pL to 100 nL, e.g., from 10 pL to 100 pL, from 10 pL to 1000 pL, from 10 pL to 10 nL, from 10 nL to 100 nL, from 1000 pL to 100 nL, from 100 pL to 100 nL, from 100 pL to 10 nL, from 100 pL to 1000 pL, from 1000 pL to 10 nL, or from 100 pL to 1000 pL. In an embodiment, the volume of the droplet is from 100 pL to 1000 pL.
In an embodiment, the droplet is a water-in-oil droplet. In an embodiment, the droplet is present in a carrier (e.g., oil) phase, e.g., a carrier phase comprising 3MTM HFE-7500 with about 1% fluorosurfactant (RAN Biotechnologies).
The droplets can be formed, e.g., using a microfluidic chip (e.g., 2R 100 pm from Dolomite) with the flow of fluid phase controlled by a syringe or pressure pump. In an embodiment, the aqueous phase of the droplet comprises a buffer, a reagent that aids cell lysis, and a bead. In an embodiment, the buffer comprises Tris at pH 7.5. In an embodiment, the reagent that aids cell lysis comprises a detergent. Exemplary detergents that can be used to aid cell lysis include, but are not limited to, Tween-20, Triton X, IGEPAL, or sodium lauroyl sarcosinate (Sarkosyl). In an embodiment, the bead is a magnetic bead. In an embodiment, the bead ses, is d to, an ucleotide (e. g., a primer), e. g., to anneal to an mRNA (e. g., an mRNA encoding a heavy chain or a light chain).
In an embodiment, the droplet contains no more than one cell after encapsulation. In an embodiment, the droplet contains a plurality of beads. In an embodiment, a plurality of beads are obtained, and at least 80%, e.g., at least 85%, 90%, 95 %, 98%, 99%, or 100%, of the ity contains no more than one cell per droplet. In an embodiment, a ity of beads are obtained, and at least 80%, e.g., at least 85%, 90%, 95%, 98%, 99%, or 100%, of the plurality contains at least one bead per droplet. Typically, the occupancy of drops is no more than one cell per droplet, and at least one bead per droplet.
Cell Lysis After encapsulation, the droplets can be incubated to facilitate cell lysis. In an embodiment, an emulsion (e. g., containing coalesce different solution phases) is heated to improved lysis efficiency in the presence of a detergent (e.g., Tween20). In an embodiment, the emulsion is incubated at a temperature between 40°C and 80°C, e. g., between 40°C and 60°C, 50°C and 70°C, or 60°C and 80°C, e. g., at 40°C, 50°C, 60°C, 70°C, or 80°C. In an embodiment, the emulsion is incubated for 5 to 60 minutes, e.g., 10 to 45 minutes, 15 to 30 minutes, 5 to 30 minutes, or 30 to 50 minutes. In an embodiment, the cell is lysed by heat. In an embodiment, the cell is lysed by an enzyme. Typically, after the cell is lysed, mRNA is released and is captured on a bead by annealing to the oligonucleotides on the bead.
Bead Recovery Emulsions (e. g., containing coalesce different solution ) can be broken using a drop destabilizing reagent, e.g., perfluorooctanol (PFO). In an embodiment, the bead-containing aqueous phase is recovered. In an embodiment, the bead is a magnetic bead, and is isolated using magnet. In an ment, the bead is washed and resuspended in a buffer (e. g., Tris, pH 7.5). In an ment, the bead is kept cold to reduce dissociation of mRNA from the bead.
Reverse Transcription Reverse transcription can be performed using rd s. In an embodiment, the reverse ription is performed in a non-emulsion reaction. In an embodiment, the reverse transcription is performed in an emulsion reaction. In a typical ment, the reverse transcription step is performed within the PCR drop. For example, mRNA-beads are encapsulated into drops with both reverse transcriptase and DNA polymerase to facilitate cDNA formation and dsDNA amplification. In an embodiment, the bead with ed mRNA is resuspended in a buffer-enzyme mix (e. g., Superscript II RT) and incubated at 35°C to 45°C (e. g., at 40°C) for 10 to 60 minutes (e.g., minutes) to facilitate e transcription. In an embodiment, the oligonucleotide coupled to the bead is used as a primer for the synthesis of the first strand cDNA. In an embodiment, the bead is washed with a buffer (e.g., Tris, pH 7.5) after reverse transcription.
Bead Encapsulationfor PCR Beads can be encapsulated individually into droplets. In an embodiment, the volume of the droplet is from 5 pL to 500 pL, e. g., from 5 pL to 400 pL, from 5 pL to 300 pL, from 5 pL to 200 pL, from 5 pL to 100 pL, from 5 pL to 50 pL, from 5 pL to 25 pL, from 400 pL to 500 pL, from 300 pL to 500 pL, from 200 pL to 500 pL, from 100 pL to 500 pL, from 50 pL to 500 pL, from 25 pL to 500 pL, from 10 pL to 500 pL, from 10 pL to 400 pL, from 25 pL to 300 pL, from 50 pL to 200 pL, or from 10 pL to 50 pL. In an embodiment, the volume of the droplet is from 10 pL to 50 pL.
In an embodiment, the t is a water-in-oil droplet. In an embodiment, the droplet is present in a carrier (e.g, oil) phase, e.g., a carrier phase comprising 3MTM HFE-7500 with about 1% fluorosurfactant (RAN Biotechnologies).
In an embodiment, the droplet contains one bead after ulation. In an embodiment, a plurality of beads are obtained, and at least 80%, e.g., at least 85%, 90%, 95%, 98%, 99%, or 100%, of the ity contains no more than one bead per droplet.
PCR Reaction In an embodiment, a PCR reaction is performed. In an embodiment, the PCR reaction is performed in a t comprising a bead that is coupled with cDNA, a DNA polymerase, oligonucleotides (e.g., for amplification of the cDNA), and a buffer.
Exemplary DNA polymerases that can be used in the reaction include, but are not limited to, Phusion® High-Fidelity DNA Polymerase (NEB), Q5® idelity DNA Polymerase (NEB), Pfu DNA polymerase, KAPA DNA polymerase, Vent® DNA polymerase, or Taq DNA polymerase.
In an embodiment, the PCR product ns an scFv cassette. In an embodiment, the scFv cassette is ucted as ker-VH. Without wishing to be bound by theory, it is believed that in an embodiment, the order can be switched to ker-VL, with no cant impact on expression or function. In an embodiment, the scFv cassette is constructed as VH-Linker-VL.
Similarly, the PCR product can contain a cassette constructed as or chain-Linker-B chain or [3 chain-Linker-a chain, or y chain-Linker-8 chain or 6 chain-Linker-y chain.
In an embodiment, a primer for a target le region sequence described herein can contain (e.g., from 5’ to 3’): a first sequence that is complementary to the sequence of an oligonucleotide attached to a capture substrate, a spacer (e.g., a spacer described herein, e.g., a PEG spacer), a ce that is complementary to at least a portion of the first sequence, a universal priming sequence, and a sequence complementary to the target variable region sequence.
In an embodiment, the reverse primer for the VL sequence contains one, two, or all of the following: (a) an overhang ce encoding a linker sequence (b) at least one modified nucleotide (e. 3., 3 consecutive nucleotides with 2’-O-methyl modification), e. g., in the ng; or (c) a 5’- phosphate. In an embodiment, the forward primer for the VH sequence contains one, two, or all of the following: (a) an overhang ce ng a linker sequence (b) at least one modified nucleotide (e.g., 3 consecutive nucleotides with 2’-O-methyl modification), e. g., in the overhang; or (c) a 5’-phosphate.
In an embodiment, the e primer for the VH sequence contains one, two, or all of the following: (a) an overhang ce encoding a linker sequence (b) at least one modified nucleotide (e. g., 3 consecutive nucleotides with 2’-O-methyl modification), e. g., in the overhang; or (c) a 5’- phosphate. In an embodiment, the forward primer for the VL sequence contains one, two, or all of the following: (a) an overhang sequence encoding a linker sequence (b) at least one modified nucleotide (e.g., 3 consecutive nucleotides with 2’-O-methyl modification), e. g., in the overhang; or (c) a 5’- phosphate.
Similarly, in an embodiment, the reverse primer for the Qt chain (or 7 chain) sequence contains one, two, or all of the ing: (a) an overhang sequence ng a linker ce (b) at least one modified nucleotide (e.g., 3 consecutive nucleotides with 2’-O-methyl modification), e. g., in the ng; or (c) a 5’-phosphate. In an embodiment, the forward primer for the B chain (or 5 chain) sequence contains one, two, or all of the following: (a) an overhang sequence encoding a linker sequence (b) at least one modified tide (e. g., 3 consecutive nucleotides with 2’-O-methyl modification), e. g., in the overhang; or (c) a 5’-phosphate.
Similarly, in an embodiment, the reverse primer for the B chain (or 5 chain) sequence contains one, two, or all of the following: (a) an overhang sequence encoding a linker sequence (b) at least one modified nucleotide (e.g., 3 consecutive nucleotides with 2’-O-methyl modification), e. g., in the overhang; or (c) a 5’-phosphate. In an ment, the forward primer for the a chain (or 7 chain) sequence contains one, two, or all of the following: (a) an ng sequence encoding a linker sequence (b) at least one modified nucleotide (e. g., 3 consecutive nucleotides with 2’-O-methyl modification), e. g., in the overhang; or (c) a 5’-phosphate.
In an embodiment, the cycling is performed with emulsion (e. g., in a PCR tube). In an embodiment, the thermocycling is med using the ing conditions: initial denaturation at 95-98°C for 30 seconds to 2 s; 10-30 cycles of: denaturation at 95-98°C for 10-30 seconds, primer annealing at 50-60°C for 10-30 seconds, and polymerase ion at 72°C for 30 seconds. In an embodiment, the reaction undergoes a slow cooling to facilitate capture of PCR products onto beads. The reaction can be hold at 4°C.
Bead Recovery Emulsions (e.g., containing coalesce different solution phases) can be broken using a drop destabilizing reagent, e. g., perfluorooctanol (PFO). In an embodiment, the bead-containing aqueous phase is recovered. In an embodiment, the bead is a magnetic bead, and is isolated using magnet. In an embodiment, the bead is washed and resuspended in a buffer (e. g., Tris, pH 7.5).
Bead Encapsulationfor Ligase Cycling Beads can be encapsulated individually into droplets. In an embodiment, the volume of the droplet is from 5 pL to 500 pL, e. g., from 5 pL to 400 pL, from 5 pL to 300 pL, from 5 pL to 200 pL, from 5 pL to 100 pL, from 5 pL to 50 pL, from 5 pL to 25 pL, from 400 pL to 500 pL, from 300 pL to 500 pL, from 200 pL to 500 pL, from 100 pL to 500 pL, from 50 pL to 500 pL, from 25 pL to 500 pL, from 10 pL to 500 pL, from 10 pL to 400 pL, from 25 pL to 300 pL, from 50 pL to 200 pL, or from 10 pL to 50 pL. In an embodiment, the volume of the droplet is from 10 pL to 50 pL.
In an ment, the droplet is a water-in-oil droplet. In an embodiment, the droplet is t in a carrier (e.g., oil) phase, e. g., a carrier phase comprising 3MTM HFE-7500 with about 1% fluorosurfactant (RAN Biotechnologies).
In an embodiment, the droplet contains one bead after encapsulation. In an embodiment, a plurality of beads are obtained, and at least 80%, e.g., at least 85%, 90%, 95%, 98%, 99%, or 100%, of the ity contains no more than one bead per droplet.
Ligase Cycling Reaction In an embodiment, a ligase cycling reaction is med. In an ment, the ligase cycling reaction is performed in a droplet comprising a bead that is coupled with PCR product, a Splint oligonucleotide (e. g., complementary and anneals to 3’ terminus of one strand (e. g., “top” VL strand) and 5’ us of another strand (e. g., “top” VH strand)), a thermostable ligase, and one or more reaction components that supports ligase enzymatic ty (e. g., NAD).
Exemplary ligases (e. g., thermostable ligases) that can be used in the reaction include, but are not limited to, Taq DNA ligase, Pfu DNA ligase, Ampligase® thermostable DNA ligase, Tsc DNA ligase, Rma DNA ligase, Tfi DNA ligase, or Tth DNA ligase.
In an embodiment, the thermocycling is performed with emulsion (e. g., in a PCR tube). In an embodiment, the thermocycling is performed using the following conditions: 3- 15 cycles of: ration at 90-95°C for 30 seconds, and annealing and ligation at 50-60°C for 1-3 minutes. The on can be hold at 4°C.
Recovery ous Portion Emulsions (e.g., containing coalesce different solution phases) can be broken using a drop destabilizing reagent, e. g., perfluorooctanol (PFO). In an embodiment, the aqueous n (e. g., containing linked product, and ally, non-linked product) is recovered. In an embodiment, the bead is discarded.
Purification ofLinked Product Linked product (e.g., representing natively linked VL—linker-VH) can be purified from non- linked products (e.g., non-linked VH and VL). The ligated products are separated from gated products by size separation. For example, denaturing PAGE (polyacrylamide gel electrophoresis), denaturing EC, agarose gel electrophoresis or AMPure XP beads can be used. The linked product (~800-900 bp) is isolated from non-linked product (~350-500 bp). For denaturing PAGE purification, the ligated band is cut out from the gel and an -elution is performed to extract DNA from gel slice (Bio-Rad Electro-Elutor).
Amplification ofPurified Linked Product The purified linked t can be amplified, e.g., by PCR. For example, the purified linked product is amplified by PCR using a DNA polymerase (e.g., Taq polymerase) under standard conditions with oligonucleotides that anneal the outer termini of the ligated product.
The final PCR product can be introduced to yeast or mammalian cells using standard methods (e.g., electroporation with expression vector) to create a natively paired library derived from ical sources.
Exemplary steps in a method of making a nucleic acid comprising a sequence that encodes a heavy chain element (HC element) of an antibody heavy chain variable region (HCVR) and a light chain element (LC element) of an antibody light chain variable region (LCVR), and n the HCVR and LCVR are matched, are illustrated in FIGS. 2A-2D.
Additional Exemplary Methods In an aspect, the sure features a method of making a nucleic acid ce comprising a sequence that encodes a heavy chain element (HC element) of an antibody heavy chain variable region (HCVR) and a light chain element (LC t) of an antibody light chain variable region (LCVR), and wherein the HCVR and LCVR are matched, the method sing carrying out the following steps from A1, B1, C1, and D1, thereby making a c acid sequence comprising a sequence that encodes an HC element of an HCVR and a LC element of an LCVR, wherein the HCVR and LCVR are matched.
In an aspect, the disclosure es a method of making a nucleic acid sequence comprising a sequence that encodes a heavy chain element (HC element) of an antibody heavy chain variable region (HCVR) and a light chain element (LC element) of an antibody light chain le region (LCVR), and wherein the HCVR and LCVR are matched, the method comprising ng out the following steps from A1, B1, C1, D2, and E1, thereby making a nucleic acid sequence comprising a sequence that encodes an HC element of an HCVR and a LC element of an LCVR, wherein the HCVR and LCVR are matched.
In an aspect, the disclosure features a method of making a c acid sequence comprising a sequence that encodes a heavy chain element (HC element) of an antibody heavy chain variable region (HCVR) and a light chain element (LC element) of an dy light chain variable region (LCVR), and wherein the HCVR and LCVR are matched, the method comprising ng out the following steps from A1, B1, C2, and D3, thereby making a nucleic acid ce comprising a ce that encodes an HC element of an HCVR and a LC element of an LCVR, n the HCVR and LCVR are matched.
In an aspect, the disclosure features a method of making a nucleic acid ce comprising a sequence that encodes a heavy chain element (HC element) of an antibody heavy chain le region (HCVR) and a light chain t (LC element) of an antibody light chain variable region (LCVR), and wherein the HCVR and LCVR are matched, the method comprising carrying out the following steps from A1, B1, C2, D4 and E2, thereby making a nucleic acid sequence comprising a ce that encodes an HC element of an HCVR and a LC element of an LCVR, wherein the HCVR and LCVR are matched.
In an aspect, the disclosure features a method of making a nucleic acid ce comprising a sequence that encodes a heavy chain element (HC element) of an dy heavy chain variable region (HCVR) and a light chain element (LC element) of an antibody light chain variable region (LCVR), and wherein the HCVR and LCVR are matched, the method comprising carrying out the following steps from A1, B1, C3, and D5, y making a nucleic acid sequence comprising a sequence that encodes an HC element of an HCVR and a LC element of an LCVR, wherein the HCVR and LCVR are matched.
In an aspect, the disclosure features a method of making a nucleic acid sequence comprising a sequence that encodes a heavy chain element (HC element) of an antibody heavy chain variable region (HCVR) and a light chain element (LC element) of an antibody light chain variable region (LCVR), and wherein the HCVR and LCVR are matched, the method comprising carrying out the following steps from A1, B1, C3, D6 and E3, thereby making a nucleic acid sequence sing a sequence that encodes an HC element of an HCVR and a LC element of an LCVR, wherein the HCVR and LCVR are matched.
In the aforesaid exemplary s, the cDNA is typically not captured on the substrate (e.g., bead) in this workflow concept. For example, mRNA dissociates from the substrate (e.g., bead), then cDNA is made in solution in the isolated reaction site (e.g., micro-chamber), e.g., in the drop, and then PCR product is made from cDNA as template in solution in the isolated reaction site (e.g., micro- chamber), e. g., in drop. In an embodiment, the method es an RT-PCR reaction, where both enzymatic steps occur in solution in drop. In the aforesaid exemplary methods, the amplified products are typically captured onto the substrate (e.g., bead), which can facilitate transitioning the paired products into the next isolated reaction site (e.g., micro-chamber), e.g., the next drop.
Antibody Molecules Disclosed herein are antibody molecules and libraries of antibody molecules. In an embodiment, the antibody molecule or library of antibody molecules are made by a method described herein.
As used herein, the term “antibody molecule” refers to a protein, e.g., an immunoglobulin chain or a fragment thereof, sing at least one imrnunoglobulin variable domain sequence. The term “antibody molecule” includes, for example, full-length, mature antibodies and antigen-binding fragments of an antibody. For example, an antibody molecule can include a heavy (H) chain variable domain sequence (abbreviated herein as VH), and a light (L) chain variable domain sequence (abbreviated herein as VL). In another example, an dy molecule includes two heavy (H) chain variable domain sequences and two light (L) chain variable domain sequence, y forming two antigen binding sites, such as Fab, Fab’, F(ab’)2, Fc, Fd, Fd’, Fv, single chain antibodies (scFv for e), single variable domain antibodies, diabodies (Dab) (bivalent and ific), and chimeric (e.g., humanized) antibodies, which may be produced by the modification of whole antibodies or those synthesized de novo using recombinant DNA technologies. These functional antibody fragments retain the ability to selectively bind with their tive antigen or receptor. Antibodies and antibody fragments can be from any class of antibodies including, but not limited to, IgG, IgA, IgM, IgD, and IgE, and from any subclass (e.g., IgGl, IgGZ, IgG3, and IgG4) of antibodies. The dy molecules can be monoclonal or polyclonal. The antibody molecule can also be a human, zed, CDR-grafted, or in vitro generated antibody. The dy molecule can have a heavy chain constant region chosen from, e.g., IgGl, IgGZ, IgG3, or IgG4. The antibody le can also have a light chain chosen from, e.g., kappa or lambda. The term “immunoglobulin” (lg) is used interchangeably with the term “antibody” herein.
Examples of antigen-binding fragments include: (i) a Fab fragment, a monovalent nt consisting of the VL, VH, CL and CH1 domains; (ii) a F(ab')2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fd nt consisting of the VH and CH1 domains; (iv) a Fv nt consisting of the VL and VH domains of a single arm of an antibody, (v) a diabody (dAb) fragment, which consists of a VH domain; (vi) a d or camelized variable domain; (vii) a single chain Fv (scFv), see e.g., Bird er al. (1988) Science 242:423-426; and Huston er al. (1988) Proc. Natl. Acad. Sci. USA 9-5883); (viii) a single domain antibody. These antibody fragments may be obtained using any suitable method, including several tional techniques known to those with skill in the art, and the fragments can be screened for utility in the same manner as are intact antibodies.
The term “antibody” includes intact les as well as functional fragments thereof.
Constant regions of the antibodies can be altered, e.g., mutated, to modify the properties of the antibody (e.3., to se or decrease one or more of: Fc receptor binding, antibody glycosylation, the number of ne residues, effector cell function, or complement function).
The antibody molecule can be a single chain antibody. A single-chain antibody (scFV) may be engineered (see, for example, Colcher, D. et al. (1999) Ann N Y Acad Sci 880:263-80; and Reiter, Y. (1996) Clin Cancer Res 2:245-52). The single chain antibody can be dimerized or multimerized to generate multivalent dies having specificities for different epitopes of the same target protein.
The antibody les disclosed herein can also be single domain antibodies. Single domain antibodies can e antibodies whose complementary determining regions are part of a single domain polypeptide. es include, but are not limited to, heavy chain antibodies, dies lly devoid of light chains, single domain antibodies derived from conventional 4- chain antibodies, engineered dies and single domain scaffolds other than those derived from antibodies. Single domain antibodies may be any of the art, or any future single domain antibodies.
Single domain antibodies may be derived from any s including, but not limited to mouse, human, camel, llama, fish, shark, goat, , and bovine. According to some aspects, a single domain antibody is a naturally occurring single domain antibody known as heavy chain antibody devoid of light chains. Such single domain antibodies are disclosed in WO 94/04678, for example.
For clarity reasons, this variable domain derived from a heavy chain antibody naturally devoid of light chain is known herein as a VHH or nanobody to distinguish it from the tional VH of four chain immunoglobulins. Such a VHH molecule can be derived from antibodies raised in Camelidae species, for example in camel, llama, dromedary, alpaca and guanaco. Other species besides Camelidae may produce heavy chain antibodies naturally devoid of light chain; such VHHs are also contemplated.
The VH and VL regions can be subdivided into regions of hypervariability, termed “complementarity determining regions” (CDR), interspersed with regions that are more conserved, termed “framework s” (FR or FW). The terms “complementarity determining region,” and “CDR,” as used herein refer to the sequences of amino acids within antibody variable s which confer n specificity and binding affinity. As used herein, the terms “framework,” “FW” and “FR” are used interchangeably.
The extent of the framework region and CDRs has been precisely defined by a number of methods (see, Kabat, E. A., et al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, US. Department of Health and Human Services, NIH Publication No. 91-3242; Chothia, C. et al. (1987) J. Mol. Biol. 196:901-917; and the AbM definition used by Oxford Molecular’s AbM antibody modeling software. See, generally, e.g., Protein Sequence and ure Analysis of Antibody Variable Domains. In: Antibody Engineering Lab Manual (Ed.: Duebel, S. and Kontermann, R., Springer-Verlag, Heidelberg). In an ment, the following definitions are used: AbM definition of CDRl of the heavy chain variable domain and Kabat definitions for the other CDRs. In an embodiment, Kabat definitions are used for all CDRs. In addition, embodiments described with respect to Kabat or AbM CDRs may also be implemented using Chothia hypervariable loops. Each VH and VL typically es three CDRs and four FRs, arranged from amino-terminus to y-terminus in the following order: FRl, CDRl, FR2, CDRZ, FR3, CDR3, and FR4.
As used herein, an “immunoglobulin variable domain sequence” refers to an amino acid sequence which can form the structure of an immunoglobulin le domain. For example, the sequence may include all or part of the amino acid sequence of a naturally-occurring variable domain.
For example, the sequence may or may not e one, two, or more N- or C-terminal amino acids, or may include other alterations that are compatible with formation of the protein structure.
The term “antigen-binding region” refers to the part of an antibody molecule that comprises determinants that form an interface that binds to an antigen, or an epitope thereof. With respect to proteins (or protein mimetics), the antigen-binding region lly includes one or more loops (of at least, e.g., four amino acids or amino acid ) that form an interface that binds to the antigen.
Typically, the antigen-binding region of an antibody molecule includes at least one or two CDRs andfor hypervariable loops, or more typically at least three, four, five or six CDRs and/or hypervariable loops.
The terms “compete” or “cross-compete” are used interchangeably herein to refer to the ability of an antibody molecule to interfere with binding of another antibody molecule, to a target.
The interference with binding can be direct or indirect (e. g., through an allosteric tion of the antibody molecule or the target). The extent to which an antibody le is able to interfere with the binding of another antibody molecule to the , and therefore whether it can be said to compete, can be ined using a competition binding assay, for example, a FACS assay, an ELISA or BIACORE assay. In an embodiment, a competition binding assay is a quantitative competition assay. In an embodiment, a first antibody molecule is said to compete for g to the target with a second antibody molecule when the binding of the first antibody molecule to the target is d by % or more, e.g., 20% or more, 30% or more, 40% or more, 50% or more, 55% or more, 60% or more, 65% or more, 70% or more, 75% or more, 80% or more, 85% or more, 90% or more, 95% or more, 98% or more, 99% or more in a competition binding assay (e.g., a competition assay described herein).
The terms “monoclonal antibody” or “monoclonal antibody composition” as used herein refer to a preparation of antibody molecules of single molecular composition. A monoclonal antibody composition displays a single binding specificity and ty for a particular e. A monoclonal antibody can be made by hybridoma technology or by methods that do not use hybridoma technology (e.g., recombinant methods).
An “effectively human” protein is a protein that does not evoke a neutralizing antibody response, e.g., the human urine antibody (HAMA) response. HAMA can be problematic in a number of circumstances, e. g., if the antibody molecule is stered repeatedly, e. g., in treatment of a chronic or recurrent disease condition. A HAMA response can make repeated antibody administration potentially ineffective because of an increased antibody nce from the serum (see, e.g., Saleh et al., Cancer Immunol. Immunother. 32:180-190 (1990)) and also because of potential allergic reactions (see, e. g., LoBuglio et at, Hybridoma, 5:5117-5123 (1986)).
The antibody molecule can be a onal or a monoclonal antibody. In some embodiments, the antibody can be inantly produced, e. g., produced by any suitable phage display or combinatorial methods.
Various phage display and combinatorial methods for generating antibodies are known in the art (as described in, e. g., Ladner et al. U.S. Patent No. 5,223,409; Kang et al. International Publication No. W0 92/18619; Dower et al. International Publication No. WO 91/17271; Winter et al.
International Publication WO 92/20791; Markland et al. International Publication No. W0 92/ 15679; Breitling et al. International Publication WO 93/01288; McCafferty et al. International Publication No. WO 92/01047; Garrard et al. International Publication No. WO 90; Ladner et al.
International Publication No. WO 90/02809; Fuchs et al. (1991) Bio/Technology 9: 1370-1372; Hay et al. (1992) Hum Antiboa’ Hybridomas 3:81-85; Huse et al. (1989) Science 246: 1275-1281; hs er al. (1993) EMBO J 12:725-734; Hawkins et al. (1992) JMol Biol 226:889-896; Clackson et al. (1991) Nature 352:624-628; Gram et al. (1992) PNAS 89:3576-3580; Garrad et al. (1991) chnology 9: 1373-1377; Hoogenboom et al. (1991) Nuc Acid Res 19:4133-4137; and Barbas et al. (1991) PNAS 88:7978-7982, the contents of all of which are incorporated by reference herein).
In an embodiment, the antibody molecule is a fully human antibody (e. g., an antibody made in a mouse which has been genetically engineered to produce an antibody from a human immunoglobulin sequence), or a man antibody, e. g., a rodent (mouse or rat), goat, e (e. g., monkey), camel antibody. In an embodiment, the non-human antibody is a rodent (mouse or rat antibody). Methods of producing rodent dies are known in the art.
Human monoclonal antibodies can be generated using transgenic mice carrying the human immunoglobulin genes rather than the mouse system. cytes from these transgenic mice immunized with the antigen of interest are used to produce hybridomas that secrete human mAbs with specific affinities for epitopes from a human n (see e. g., Wood et al. ational Application WO 06, Kucherlapati et al. PCT publication W0 91/ 10741; Lonberg et al. International Application WO 92/03918; Kay et al. International Application 92/03917; Lonberg, N. et al. 1994 Nature 368:856-859; Green, L.L. et al. 1994 Nature Genet. 7:13-21; on, S.L. et al. 1994 Proc.
Natl. Acad. Sci. USA 1-6855; Bruggeman et al. 1993 Year Immunol 7:33-40; Tuaillon et al. 1993 PNAS 90:3720—3724; Bruggeman et al. 1991 Eur J l 21:1323-1326).
An antibody can be one in which the variable , or a portion thereof, e. g., the CDRs, are generated in a man organism, e.g., a rat or mouse. Chimeric, CDR-grafted, and humanized antibodies are within the invention. Antibodies generated in a non-human organism, e. g., a rat or mouse, and then modified, e.g., in the variable framework or constant , to decrease antigenicity in a human are within the invention.
Chimeric antibodies can be produced by any suitable recombinant DNA technique. Several are known in the art (see Robinson et al., International Patent Publication PCT/US86/02269; Akira, et al., European Patent Application 7; Taniguchi, M., European Patent Application 171,496; Morrison et al., European Patent Application 4; Neuberger et al., International Application WO 86301533; Cabilly et al. US. Patent No. 4,816,567; Cabilly et al., European Patent Application 125,023; Better et al. (1988 Science 240: 1041-1043); Liu et al. (1987) PNAS 9-3443; Liu et al., 1987, J. l. 139:3521-3526; Sun et al. (1987) PNAS 84:214-218; Nishimura et al., 1987, Canc. Res. 47:999-1005; Wood et al. (1985) Nature 314:446—449; and Shaw et al., 1988, J. Natl Cancer Inst. 80: 1553- 1559).
A humanized or CDR-grafted antibody will have at least one or two but generally all three recipient CDRs (of heavy and or light globulin chains) replaced with a donor CDR. The antibody may be replaced with at least a portion of a non-human CDR or only some of the CDRs may be replaced with man CDRs. It is only ary to replace the number of CDRs required for binding of the humanized antibody to an antigen. In an embodiment, the donor will be a rodent antibody, e. g., a rat or mouse antibody, and the recipient will be a human framework or a human consensus framework. Typically, the globulin providing the CDRs is called the “donor” and the immunoglobulin providing the framework is called the tor.” In some embodiments, the donor immunoglobulin is a non-human (e.g., rodent). The acceptor framework is typically a naturally-occurring (e.g., a human) framework or a consensus framework, or a sequence about 85% or higher, e.g., 90%, 95%, 99% or higher identical thereto.
As used herein, the term nsus sequence” refers to the sequence formed from the most frequently occurring amino acids (or tides) in a family of related sequences (See e.g., Winnaker, From Genes to Clones (Verlagsgesellschaft, Weinheim, Germany 1987). In a family of proteins, each position in the sus sequence is occupied by the amino acid occurring most frequently at that on in the family. If two amino acids occur y frequently, either can be included in the consensus sequence. A “consensus framework” refers to the framework region in the consensus immunoglobulin sequence.
An antibody can be humanized by any suitable method, and several such s known in the art (see e.g., Morrison, S. L., 1985, Science 229: 1202-1207, by Oi et al., 1986, BioTechniqaes 4:214, and by Queen et al. US 5,585,089, US 5,693,761 and US 5,693,762, the contents of all of which are hereby incorporated by reference).
Humanized or CDR-grafted antibodies can be produced by CDR-grafting or CDR substitution, wherein one, two, or all CDRs of an immunoglobulin chain can be replaced. See e.g., US. Patent 5,225,539; Jones et al. 1986 Nature 321 :552-525; Verhoeyan et al. 1988 Science 239:1534; r et al. 1988 J. Immunol. 141:4053-4060; Winter US 5,225,539, the contents of all of which are hereby expressly incorporated by reference. Winter describes a CDR-grafting method which may be used to prepare humanized antibodies (UK Patent Application GB 2188638A, filed on March 26, 1987; Winter US 5,225,539), the contents of which is expressly incorporated by nce.
Also provided are humanized antibodies in which specific amino acids have been tuted, deleted or added. Criteria for selecting amino acids from the donor are bed in, e. g., US ,585,089, e.g., s 12-16 of US 5,585,089, the contents of which are hereby incorporated by nce. Other techniques for humanizing antibodies are described in Padlan et al. EP 519596 A1, published on December 23, 1992.
In an embodiment, the dy le has a heavy chain constant region chosen from, e.g., the heavy chain constant regions of IgG1, IgG2 (e.g., IgG2a), IgG3, IgG4, IgM, IgAl, IgA2, IgD, and IgE; particularly, chosen from, e.g., the (e.g., human) heavy chain constant regions of IgG1, IgG2, IgG3, and IgG4. In another embodiment, the antibody molecule has a light chain constant region chosen from, e. g., the (e.g., human) light chain constant regions of kappa or lambda. The constant region can be altered, e. g., mutated, to modify the properties of the dy molecule (e.g., to increase or decrease one or more of: Fc receptor g, antibody glycosylation, the number of cysteine residues, effector cell function, and/or complement on). In an embodiment, the dy molecule has or function and can fix complement. In another embodiment, the antibody molecule does not recruit effector cells or fix complement. In certain embodiments, the antibody molecule has reduced or no ability to bind an Fc receptor. For example, it may be an isotype or subtype, fragment or other mutant, which does not support binding to an Fc or, e. g., it has a mutagenized or deleted Fc receptor binding region.
In an embodiment, a constant region of the antibody molecule is altered. Methods for altering an antibody constant region are known in the art. dy molecules with altered function, e.g. altered affinity for an effector ligand, such as FcR on a cell, or the C1 component of complement can be produced by replacing at least one amino acid residue in the constant portion of the antibody with a different residue (see e.g., EP 388,151 A1, US. Pat. No. 5,624,821 and US. Pat. No. 5,648,260, the contents of all of which are hereby incorporated by reference). Amino acid mutations which stabilize antibody structure, such as S228P (EU nomenclature, S241P in Kabat nomenclature) in human IgG4 are also contemplated. Similar type of alterations could be described which if applied to the murine, or other species immunoglobulin would reduce or ate these functions.
In an embodiment, the only amino acids in the antibody molecule are canonical amino acids.
In an embodiment, the antibody le comprises naturally-occurring amino acids; analogs, derivatives and congeners thereof; amino acid analogs having variant side chains; and/or all stereoisomers of any of any of the foregoing. The antibody molecule may comprise the D- or L- optical s of amino acids and peptidomimetics.
A polypeptide of an antibody molecule described herein may be linear or branched, it may comprise modified amino acids, and it may be interrupted by non-amino acids. The antibody WO 19402 molecule may also be modified; for example, by disulfide bond ion, glycosylation, lipidation, acetylation, phosphorylation, or any other manipulation, such as conjugation with a labeling component. The polypeptide can be isolated from natural sources, can be a produced by recombinant techniques from a otic or prokaryotic host, or can be a product of synthetic procedures.
The antibody molecule bed herein can be used alone in unconjugated form, or can be bound to a substance, e. g., a toxin or moiety (e.g., a therapeutic drug; a compound emitting radiation; molecules of plant, fungal, or bacterial origin; or a biological protein (e.g., a protein toxin) or particle (e.g., a recombinant Viral particle, e.g., Via a Viral coat protein). For example, the antibody molecule can be coupled to a radioactive isotope such as an (1-, [3-, or y-emitter, or a B-and y-emitter.
An antibody molecule can be derivatized or linked to another functional molecule (e.g., another peptide or protein). As used herein, a “derivatized” antibody molecule is one that has been modified. Methods of derivatization include but are not limited to the addition of a cent moiety, a radionucleotide, a toxin, an enzyme or an affinity ligand such as biotin. Accordingly, the dy les are intended to include derivatized and otherwise modified forms of the antibodies described herein, including immunoadhesion molecules. For example, an antibody molecule can be functionally linked (by al coupling, genetic fusion, noncovalent association or otherwise) to one or more other molecular entities, such as r antibody (e.g., a bispecific antibody or a diabody), a detectable agent, a toxin, a pharmaceutical agent, andior a n or e that can mediate association of the antibody or antibody portion with another molecule (such as a avidin core region or a polyhistidine tag).
Some types of tized antibody molecule are produced by crosslinking two or more antibodies (of the same type or of different types, e.g., to create bispecific antibodies). Suitable crosslinkers include those that are heterobifunctional, having two distinctly reactive groups separated by an appropriate spacer (e.g., m-maleimidobenzoyl-N-hydroxysuccinimide ester) or homobifunctional (e.g., disuccinimidyl suberate). Such s are ble from Pierce Chemical Company, Rockford, Ill.
Useful detectable agents with which an antibody molecule may be derivatized (or d) to include cent compounds, various enzymes, prosthetic groups, luminescent materials, bioluminescent materials, fluorescent emitting metal atoms, e.g., europium (Eu), and other anthanides, and radioactive materials (described below). Exemplary cent detectable agents include cein, fluorescein isothiocyanate, rhodamine, 5dimethylamine-l-napthalenesulfonyl chloride, phycoerythrin and the like. An antibody may also be derivatized with detectable enzymes, such as alkaline phosphatase, horseradish peroxidase, B-galactosidase, acetylcholinesterase, glucose oxidase and the like. When an antibody is derivatized with a detectable enzyme, it is detected by adding additional reagents that the enzyme uses to produce a detectable reaction product. For example, when the detectable agent horseradish peroxidase is present, the addition of en peroxide and diaminobenzidine leads to a colored reaction product, which is detectable. An antibody molecule may also be tized with a etic group (e.g., streptavidin/biotin and avidin/biotin). For example, an antibody may be derivatized with biotin, and detected through indirect measurement of avidin or streptavidin binding. Examples of suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin; an example of a luminescent material includes luminol; and examples of bioluminescent materials include luciferase, luciferin, and aequorin.
Labeled antibody molecules can be used, for example, diagnostically and/or experimentally in a number of contexts, ing (i) to isolate a predetermined antigen by standard techniques, such as affinity tography or immunoprecipitation; (ii) to detect a predetermined antigen (e.g., in a cellular lysate or cell supernatant) in order to evaluate the abundance and pattern of expression of the protein; (iii) to monitor protein levels in tissue as part of a clinical testing procedure, e.g., to determine the efficacy of a given treatment regimen.
An antibody molecule may be conjugated to another lar entity, typically a label or a therapeutic (e.g., crobial (e. g., antibacterial or bactericidal), immunomodulatory, immunostimularoty, cytotoxic, or cytostatic) agent or moiety. Radioactive isotopes can be used in diagnostic or therapeutic applications. Radioactive es that can be coupled to the antibody les include, but are not limited to or-, [3-, or y—emitters, or B-and y—emitters. Such radioactive es include, but are not limited to iodine (1311 or 125I), yttrium (90Y), lutetium (177Lu), um (225Ac), praseodymium, astatine (leAt), rhenium (186Re), bismuth (212Bi or 213Bi), indium (1 1 1In), technetium (gngc), phosphorus (32F), rhodium (lgth), sulfur (35S) carbon (14C), tritium (3H), chromium (5 lCr), chlorine (36Cl), cobalt (57Co or 58C0), iron (59Fe), selenium (75Se), or gallium (67Ga).
Radioisotopes useful as therapeutic agents include m (90Y), lutetium (177Lu), actinium (225Ac), praseodymium, astatine (leAt), rhenium (186Re), bismuth (212Bi or 213Bi), and rhodium (lgth).
Radioisotopes useful as labels, e.g., for use in diagnostics, e iodine (1311 or 125I), indium 1), technetium ), phosphorus (32F), carbon (14C), and tritium (3H), or one or more of the therapeutic isotopes listed above.
The present disclosure provides radiolabeled antibody molecules and methods of labeling the same. In an embodiment, a method of labeling an antibody molecule is disclosed. The method includes ting an antibody molecule, with a ing agent, to thereby produce a conjugated antibody. The conjugated antibody is radiolabeled with a sotope, e.g., 111Indium, 90Yttrium and 177Lutetium, to thereby produce a labeled antibody molecule.
In some aspects, this disclosure provides a method of making an antibody molecule disclosed herein. The method includes: providing an antigen, or a fragment thereof; obtaining an antibody molecule that specifically binds to the antigen; ting efficacy of the antibody molecule in ting activity of the antigen and/or organism expressing the antigen. The method can further e administering the antibody molecule, including a derivative thereof (6.3., a humanized antibody molecule) to a subject, e.g., a human.
This disclosure provides an isolated nucleic acid molecule encoding the above antibody molecule, vectors and host cells thereof. The nucleic acid molecule includes, but is not limited to, RNA, genomic DNA and cDNA.
Other Binding Polypeptides The disclosures herein are not intended to be limited to antibody molecules. The methods described herein are broadly applicable to any binding polypeptides that have two or more chains (e.g., having at least two paired or matched chains).
In an embodiment, the g molecule comprises an X chain variable region and a Y chain variable region. For example, in any of the aspects, embodiments, and definitions herein, an dy heavy chain (or variable region) can be replaced with an X chain (or variable ), and an antibody light chain (or le region) can be replaced with a Y chain (or variable region).
In an aspect, the disclosure features a method of making a nucleic acid sequence comprising a sequence that encodes an X chain element (XC element) of an antibody heavy chain variable region (XCVR) and a Y chain t (YC element) of an antibody light chain variable region (YCVR), and wherein the XCVR and YCVR are matched, the method comprising: a) acquiring an isolated production reaction site, e.g., a production micro-chamber, comprising: i) an X chain (XC) strand, n the XC strand is a strand of an X chain -stranded cDNA (XC ds cDNA) comprising a t that encodes an XC element of the XCVR from a cell, e.g., an X chain variable region sequence (XCVRS); and ii) a Y chain (YC) strand, wherein the YC strand is a strand of a Y chain double-stranded cDNA (YC ds cDNA) comprising a segment that s a YC element of the YCVR from the cell, e.g., a Y chain variable region sequence (YCVRS), and b) nt linking, e.g., ligation, of an XC strand to a YC strand, wherein the isolated production reaction site, e.g., a production micro-chamber, does not include a nucleic acid encoding a YCVR or an XCVR from a cell other than the cell (e.g., a ent cell), thereby making a nucleic acid sequence comprising a sequence that encodes an XC element of an XCVR and a YC element of a YCVR, wherein the XCVR and YCVR are matched.
“Matched,” as that term is used herein in connection with an X chain variable region and a Y chain variable region, means they are from the same cell. In an embodiment, the X chain variable region and the Y chain variable region can form a multimeric protein or a part of a multimeric protein.
With respect to an t of an X chain variable region and an element of a Y chain variable region it means that the X chain variable region and the Y chain le region from which the ts are derived are from the same cell.
An “X chain variable region sequence,” or “XCVRS,” as that term is used herein, refers to a polypeptide comprising sufficient ce to allow binding of another polypeptide (e.g., an antigen).
In embodiments the XCVRS can assemble with a Y chain variable region, and, e.g., bind antigen. A “Y chain variable region sequence,” or “YCVRS,” as that term is used herein, refers to a polypeptide comprising ent sequence to allow binding of another polypeptide (e.g., an antigen). In embodiments the YCVRS can assemble with an X chain variable region, and, e.g., bind antigen.
“Element” of an XC or YC le region, as that term is used herein, refers to a sequence that encodes at least one amino acid. In an embodiment, an element comprises a CDR. In an embodiment an element comprises a FW region.
In an ment, the XC element comprises, or consists of, an XCVRS. In an embodiment, the YC element comprises, or consists of, a YCVRS.
In an embodiment, the XC ds cDNA comprises a segment that encodes an XCVRS. In an embodiment, the YC ds cDNA ses a segment that encodes a YCVRS. In an embodiment, the XC ds cDNA comprises a segment that encodes an XCVRS, and the YC ds cDNA comprises a t that encodes a YCVRS.
In an embodiment, the cell is an immune cell, e.g., a B cell, e.g., a human B cell. In an embodiment, the cell is a mammalian cell or an avian cell.
In an embodiment, the nucleic acid sequence is configured such that, when expressed, the XC element and the YC element (e.g., the XCVRS and the YCVRS) form a functional n binding molecule, e. g., a single chain or a complex of an XC and a YC. In an embodiment, the n binding molecule is functional in vitro, ex vivo, or in vivo, e.g., as determined by a method or assay described herein.
In an embodiment, acquiring an isolated production reaction site, e.g., a production micro- chamber, comprises: a) acquiring a capture substrate bound to: (i) a first double-stranded cDNA (ds cDNA) comprising a strand that is complementary to a first mRNA that encodes an XCVR from a cell; and (ii) a second ds cDNA sing a strand complementary to a second mRNA encoding a YCVR from the cell (the cDNA loaded capture substrate), and b) maintaining the ed production reaction site, e.g., the production micro-chamber, under conditions that allow amplification of the first and second ds cDNAs, to produce: a plurality of XC ds cDNAs comprising a segment that encodes an XC element of the XCVR from the cell, e.g., an XCVRS; and a ity of YC ds cDNAs comprising a segment that encodes a YC element of the YCVR from the cell, e.g., a YCVRS.
In an ment, the XC ds cDNA is identical, or substantially identical, to the first ds cDNA. For example, the sense strand of the XC ds cDNA is at least 80%, 85%, 90%, 95%, 98%, 99%, or 100% identical to, or differs by no more than 1, 2, 5, 10, 15, 20, 25, 30, 35, 40, 45, or 50 nucleotides from, the sense strand of the first ds cDNA, and/or the antisense strand of the XC ds cDNA is at least 80%, 85%, 90%, 95%, 98%, 99%, or 100% identical to, or differs by no more than 1, 2, 5, 10, 15, 20, 25, 30, 35, 40, 45, or 50 nucleotides from, the antisense strand of the first ds cDNA.
In an embodiment, the YC ds cDNA is cal, or substantially identical, to the second ds cDNA. For example, the sense strand of the YC ds cDNA is at least 80%, 85 %, 90%, 95%, 98%, 99%, or 100% identical to, or differs by no more than 1, 2, 5, 10, 15, 20, 25, 30, 35, 40, 45, or 50 nucleotides from, the sense strand of the second ds cDNA, and/or the antisense strand of the YC ds cDNA is at least 80%, 85%, 90%, 95%, 98%, 99%, or 100% identical to, or differs by no more than 1, 2, 5, 10, 15, 20, 25, 30, 35, 40, 45 , or 50 nucleotides from, the antisense strand of the second ds cDNA.
In an embodiment, the XC strand is a sense strand. In an embodiment, the YC strand is a sense strand. In an embodiment, the XC strand is an antisense strand. In an embodiment, the YC strand is an antisense . In an ment, both the XC strand and the YC strand are sense strands. In an embodiment, both the XC strand and the YC strand are antisense strands.
In an embodiment, the capture substrate comprises a bead, e.g., a magnetic bead. In an ment, the capture substrate comprises a moiety (e.g., an oligonucleotide) which binds to cDNA, e.g., (i) a moiety which binds to the XC strand; (ii) a moiety which binds to the YC strand; or (iii) both (i) and (ii). In an embodiment, the moiety which binds to the XC strand is ent from the moiety which binds to the YC strand, e.g., to facilitate creating conditions favorable to capturing similar levels of each DNA molecule type. In an embodiment, the moiety which binds to the XC strand is identical to the moiety which binds to the YC strand.
In an embodiment, the first mRNA and the second mRNA are disposed on an mRNA loaded capture substrate.
In an embodiment, the isolated production reaction site, e.g., the production chamber, comprises: a reagent mixture suitable for producing, from the first and second mRNAs (e.g., after the first and second mRNAs are released from the mRNA loaded capture substrate into a solution), a first cDNA comprising a segment that s an XC t of the XCVR of the cell, e.g., an XCVRS, and a second cDNA comprising a segment that encodes a YC element of the YCVR of the cell, e.g., a YCVRS.
In an embodiment, the isolated production reaction site, e.g., production micro-chamber, comprises primers that mediate the production of the first ds cDNA. In an embodiment, the isolated production reaction site, e.g., production chamber, comprises primers that mediate the production of the second ds cDNA.
In an embodiment, a cDNA strand that is complementary to a first mRNA that s an XCVR from a cell is made by reverse transcription of the first mRNA. In an embodiment, a cDNA strand that is complementary to a second mRNA that s a YCVR from a cell is made by reverse transcription of the second mRNA.
In an embodiment, the reverse transcription takes place in the isolated production reaction site, e.g., a production-micro chamber. In an embodiment, the reverse ription takes place in an isolated cell reaction site, e. g., a cell isolation micro-chamber. In an embodiment, the reverse transcription takes place outside the isolated production reaction site, e. g., a production micro- chamber, or outside an ed cell reaction site, e. g., a cell isolation micro-chamber. In an embodiment, the reverse transcription takes place e the isolated tion reaction site, e.g., a production-micro chamber, and e an isolated cell reaction site, e.g., a cell isolation micro- chamber. In an embodiment, the reverse transcription takes place outside an isolated reaction site, e.g., outside a micro-chamber.
In an embodiment, the amplification comprises 30 or fewer cycles, e.g., 20 or fewer cycles, e.g., 15 or fewer, 14 or fewer, 13 or fewer, 12 or fewer, 11 or fewer, 10 or fewer, 9 or fewer, 8 or fewer, 7 or fewer, 6 or fewer, or 5 or fewer cycles.
In an ment, the reverse transcription and/or amplification uses one or more primers, e. g., comprising a sequence specific for an XCVRS and/or a YCVRS.
In an embodiment, the reverse ription and/or amplification comprises using two or more primers that e the production of the XC ds cDNA, wherein at least one primer comprises a nucleotide modification, and wherein at least one primer does not se a nucleotide modification.
In an embodiment, the amplification comprises using two or more primers that mediate the production of the YC ds cDNA, wherein at least one primer comprises a nucleotide modification, and n at least one primer does not comprise a nucleotide modification.
In an embodiment, at least one primer ses a nucleotide modification, e.g., which reduces, e. g., ts, DNA synthesis, e.g., by a DNA polymerase. In an embodiment, at least one primer does not comprise a nucleotide modification, e.g., which reduces, e.g., inhibits, DNA synthesis, 6.3., by a DNA polymerase.
In an embodiment, the nucleotide ation inhibits a DNA polymerase from extending the DNA. Without wishing to be bound by theory, it is believed that in an embodiment any al entity that reduces (e.g., blocks) DNA polymerase extension can be used in accordance with the methods bed herein.
In an embodiment, the nucleotide modification is an insertion of a spacer to the primer, e. g., between two adjacent nucleotides in the primer. In an embodiment, the spacer is a flexible spacer. In an embodiment, the spacer is a carbon spacer (e.g., n-, wherein n=3, 4, 5, 6, 7, 8, 9, 10, or more), two or more (e.g., three, four, five, six, seven, eight, nine, ten, or more) abasic nucleotides, or a polyethylene glycol (PEG) spacer. In an embodiment, the spacer is a PEG spacer. In an embodiment, the nucleotide ation is 2’-O-methyl, 2’-OH, 2’-NH2, or uracil, e. g., to a ribose.
In an embodiment, the nucleotide modification is located internally or at the 3’ end of the primer. In an embodiment, at least one primer comprises (i) a first member; (ii) a second member; and optionally (iii) a third member, e.g., comprising a nucleotide modification described herein, e.g., located between (i) and (ii).
In an embodiment, the first member is capable of annealing with the second member. In an embodiment, the first member is capable of ing with the second member in the same primer, e.g., through intra-molecular hybridization, e.g., to form a hairpin structure comprising a duplex region of 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, more irs. In another embodiment, the first member is capable of annealing hybridizing with the second member in a different primer, e.g., through inter-molecular hybridization, e. g., to form a double-stranded structure comprising a duplex region of 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, more basepairs. Without wishing to be bound by theory, it is believed that in an ment, there are at least two secondary structures that the modified primers can form and facilitate reduction (e.g., prevention) of competition to ate (e.g., bead) capture. For example, the secondary structure can be a hairpin-like structure formed by intra-molecular hybridization (within the same primer), or the secondary structure can be a duplex structure formed by inter-molecular hybridization (between two different primers).
In an embodiment, the first member comprises a ce that is mentary to the sequence of an ucleotide attached to the capture ate. In an ment, the second member comprises (e.g., from 5’ to 3’) one, two, or all of: (i) a sequence that is complementary to at least a portion of the first member; (ii) a universal priming ce (e.g., for PCR amplification or next-generation sequencing); and (iii) a sequence complementary to a target sequence, e. g., an XCVRS and/or a YCVRS. In an embodiment, the universal priming sequence is identical, or substantially identical, to the sequence that is mentary to at least a portion of the first member.
In another embodiment, the universal priming sequence is different from the sequence that is complementary to at least a portion of the first member. In an embodiment, the second member comprises a sequence for homologous recombination (e.g., in a yeast or mammalian cell).
In an ment, at least one primer comprises a sequence encoding at least a portion of a linker sequence, or a mentary sequence thereof. In an embodiment, the primer that comprises a sequence encoding at least a portion of a linker sequence, or a complementary sequence thereof, is orylated, e.g., 5 ’ phosphorylated. Without g to be bound by theory, it is believed that in an embodiment, any ce with the general properties of flexibility (e.g., facilitated by glycine) and hydrophilicity can work effectively in accordance with the methods described herein. Exemplary linkers can generally have overrepresentation of one or more of Gly, Ser, Thr, or Ala and underrepresentation of hydrophobic residues, e.g., one or more of Trp, Tyr, Phe, Cys, Met, Leu, or Ile.
The length of the primer may vary, e.g., 3-50 amino acid residues (e.g., 5-45, 10-40, 15-35, 20-30, 10- 20, 10-30, 20-40, or 30-40 amino acid residues). In an embodiment, the linker sequence comprises, or consists of, ((Gly)m-Ser))n, where m=3, 4, 5, or more and n=1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more. In an embodiment, the linker sequence comprises, or consists of, (Gly-Gly-Gly-Gly-Ser)n, where n=1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more.
In an embodiment, the primer is a primer described , e.g., in Examples.
In an embodiment, the reverse ription, the cation, or both, occurs in a solution in the isolated production reaction site, e. g., production micro-chamber. In an embodiment, the e transcription, the amplification, or both, does not occur on the substrate (e.g., bead). For example, the e transcription, the amplification, or both, can occur on in a solution within a droplet.
In an ment, the XC ds cDNA comprises a 5’ overhang, e.g., a 5’ overhang that is capable of hybridizing to an oligonucleotide attached to a capture substrate. In an embodiment, the KC ds cDNA comprises a blunt end, e.g., a blunt end comprising a 5’ phosphate. In an embodiment, the YC ds cDNA comprises a 5’ overhang, e.g., a 5’ overhang that is capable of hybridizing to an oligonucleotide attached to a capture substrate. In an embodiment, the YC ds cDNA comprises a blunt end, 6.3., a blunt end comprising a 5’ phosphate. In an embodiment, the XC ds cDNA and the YC ds cDNA comprise sticky ends, e.g., both have 5’ overhangs.
In an embodiment, the XC strand and the YC strand are covalently linked, e.g., ligated, to produce a single stranded nucleic acid sequence, wherein the XC and YC strands are both sense strands or both antisense strands. In an embodiment, a denatured XC strand of the XC ds cDNA to a denatured YC strand of the YC ds cDNA are covalently linked, e. g., ligated, wherein the XC and YC s are both sense strands or both antisense strands. In an embodiment, the XC strand is present in the XC ds cDNA and the YC strand is present in the YC ds cDNA, and wherein the XC ds cDNA and the YC ds cDNA are covalently linked, e.g., ligated, e.g., to produce a double stranded nucleic acid sequence.
In an embodiment, the covalent linking, e.g., ligation, occurs in the isolated tion reaction site. In an embodiment, the isolated tion reaction site, e.g., a tion micro- chamber, or the isolated linkage reaction site, e.g., a e micro-chamber, comprises a reagent that is capable of covalently linking, e.g., ng, the XC and YC strands or the XC and YC ds cDNAs.
In an embodiment, the isolated production on site, e.g., a production micro-chamber comprises an enzyme that covalently couples the XC and YC s or the XC and YC ds cDNAs. In an embodiment, the enzyme is a ligase, e.g., a thermal stable ligase. In an embodiment, the covalent linking ses ligase thermocycling.
In an embodiment, the covalent linking, e.g., on, occurs in a site different from the isolated production on site, e.g., occurs in an isolated linkage reaction site, e.g., a linkage micro- chamber. In an embodiment, the XC strand and the YC strand are transferred from the isolated production site to the isolated linkage reaction site, e.g., a linkage micro-chamber, and the covalent linking occurs in the isolated e reaction site, e.g., a linkage micro-chamber. In an embodiment, the isolated linkage reaction site, e.g., a linkage micro-chamber, comprises a reagent that is capable of covalently linking, e.g., ligating, the XC and YC strands or the XC and YC ds cDNAs. In an embodiment, the ed linkage reaction site, e.g., a linkage micro-chamber, comprises an enzyme that covalently couples the XC and YC s or the XC and YC ds cDNAs. In an embodiment, the enzyme is a ligase, e.g., a thermal stable ligase. In an embodiment, the nt linking ses ligase thermocycling.
In an ment, the covalent linking, e. g., ligation, comprises: (a) g the isolated linkage reaction site, e.g., the linkage micro-chamber, under conditions (e.g., at 950C) that allow ration of the XC strand and the YC strand; (b) cooling the isolated linkage reaction site, e. g., the e micro-chamber, under ions (e.g., at 50-65°C) that allow hybridization of the splint oligonucleotide to the XC strand and the YC strand; (c) maintaining the isolated linkage reaction site, e.g., the linkage micro-chamber, under conditions (e.g., at 45-65°C) that allow ligation of the XC strand and the YC strand (e.g., formation of odiester bond between the XC strand and the YC strand); and (d) repeating steps (a), (b), and (c) tially for 2, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, or more cycles.
In an embodiment, the XC strand and the YC strand are covalently linked, e.g., ligated, in the presence of a splint oligonucleotide. In an embodiment, the splint oligonucleotide is hybridized to a sequence comprising the junction of the XC strand and the YC strand, or a sequence complementary thereof, and forms a duplexed region at the site of ligation. In an embodiment, the splint oligonucleotide comprises a modification (e.g., an NHZ group) that inhibits DNA synthesis, e.g., by a DNA polymerase. In an embodiment, the modification is at the 3’ end of the splint oligonucleotide.
In an embodiment, a strand complimentary to the covalently linked, e. g., ligated, XC and YC strands is produced by amplification.
In an ment, the method, e.g., the step of covalent linkage, does not include a step of overlap extension polymerase chain reaction (OE-PCR), also known as splicing by overlap ion or splicing by overhang extension (SOE) PCR.
In an embodiment, the method further comprises, prior to acquiring the isolated production reaction site, e.g., a production micro-chamber, acquiring an mRNA loaded capture ate.
In an embodiment, ing the mRNA loaded capture substrate comprising: a) acquiring an isolated cell reaction site, e.g., a cell isolation micro-chamber, comprising: i) a cell; and ii) a capture substrate capable of binding a first mRNA encoding an XCVR from the cell and a second mRNA encoding a YCVR from the cell; and b) maintaining the isolated cell reaction site, e.g., the cell isolation micro-chamber, under conditions that allow lysis of the cell and binding of the capture substrate with the first mRNA and the second mRNA to form the mRNA loaded capture substrate, wherein the isolated cell reaction site, e.g., cell isolation micro-chamber, does not include a c acid encoding an XCVR or a YCVR from a cell other than the cell (e.g., a different cell).
In an embodiment, the isolated cell reaction site, e. g., cell isolation micro-chamber, comprises a lysing reagent, e.g., a detergent. In an embodiment, the cell is lysed by heat or an enzyme. In an embodiment, the capture substrate ses a moiety (e.g., an oligonucleotide) which binds mRNA, e.g., an oligo(dT).
In an embodiment, the method further comprises releasing the mRNA loaded capture substrate from the isolated cell reaction site, e.g., the cell isolation micro-chamber. In an embodiment, the releasing step is performed in the presence of a poly(dA) or poly(dT) oligonucleotide, e.g., to reduce binding of non-captured mRNA.
In an embodiment, the mRNA loaded capture ate is transferred from the isolated cell reaction site, e.g., the cell isolation micro-chamber, to the ed production reaction site, e.g., the tion micro-chamber.
In an ment, the method further ses releasing the nucleic acid sequence from the isolated production reaction site, e.g., the production micro-chamber. In an embodiment, the method further comprises amplifying the nucleic acid sequence. In an embodiment, amplification of the nucleic acid sequence occurs outside the isolated production reaction site, e.g., the production micro- chamber, e. g., after the nucleic acid is ed from the ed production reaction site, e. g., the tion micro-chamber. In an embodiment, amplification of the nucleic acid sequence occurs at the isolated production on site, e.g., the production micro-chamber.
In an embodiment, the method further comprises sequencing all or a portion of the c acid sequence.
In an embodiment, the method further ses inserting all or a portion of nucleic acid sequence into a vector. In an embodiment, the vector supplies an additional XC element or YC element not included in the nucleic acid sequence. In an embodiment, the vector supplies an XC CDRl, an XC CDR2, or both. In an embodiment, the method further comprises sing the vector.
In an embodiment, the method further comprises expressing the nucleic acid sequence to produce a polypeptide comprising a segment that encodes an XC element of the XCVR, e.g., an XCVRS, and a segment that encodes a YC element of the YCVR, e.g., a YCVRS. In an embodiment, the YC element is N—terminal to the KC element in the polypeptide. In an embodiment, the XC element is C-terminal to the YC element in the polypeptide.
In an embodiment, the method further comprises contacting the polypeptide with an antigen.
In an embodiment, the method further comprises determining if the polypeptide binds the n, in vitro, ex vivo, or in viva, e.g., by a method or assay described herein.
In an aspect, the disclosure features a method of making a nucleic acid sequence comprising a sequence that encodes an X chain element (XC t) of an antibody heavy chain variable region (XCVR) and a Y chain element (YC element) of an antibody light chain variable region (YCVR), and wherein the XCVR and YCVR are matched, comprising: a) acquiring an isolated cell reaction site (e.g., an isolated cell reaction site bed herein), e.g., a cell isolation micro-chamber, comprising: i) a cell (e.g., a cell described herein); and ii) a capture substrate (e. g., a capture substrate described herein) capable of g a first mRNA ng an XCVR from the cell and a second mRNA encoding a YCVR from the cell; b) maintaining the isolated cell reaction site, e.g., the cell isolation micro-chamber, under conditions that allow lysis of the cell and binding of the capture substrate with the first mRNA and the second mRNA to form an mRNA loaded capture substrate, wherein the isolated cell on site, e.g., cell isolation chamber, does not include a nucleic acid encoding an XCVR or a YCVR from a cell other than the cell (e.g., a different cell); c) contacting the mRNA loaded capture substrate with a reaction mixture, e.g., a reaction mixture sing reverse transcriptase, that uses the loaded mRNA as a template to make cDNA (this can occur, e.g., in the isolated cell reaction site, in an isolated production reaction site, or in neither, e.g., not in an isolated on site); (1) acquiring an isolated production reaction site (e.g., an isolated production reaction site described herein), e.g., a production chamber, comprising: i) an X chain (XC) strand, n the XC strand is a strand of an X chain double-stranded cDNA (XC ds cDNA) comprising a t that encodes an XC element of the XCVR from the cell, e.g., an X chain variable region sequence (XCVRS); and ii) a Y chain (YC) strand, wherein the YC strand is a strand of a Y chain double- stranded cDNA (YC ds cDNA) comprising a segment that encodes a YC element of the YCVR from the cell, e.g., a Y chain variable region sequence (YCVRS), wherein the isolated tion on site, e.g., a production micro-chamber, does not e a nucleic acid encoding a YCVR or an XCVR from a cell other than the cell (e. g., a different cell); and e) covalent linking, e. g., ligation, of the XC strand to the YC strand.
In an embodiment, one or more (e.g., two, three, four, or all) of the steps a)-e) are performed in accordance with a method described herein. In an embodiment, each of the steps a)-e) is performed in accordance with a method described herein.
In an , the disclosure features a method of making a nucleic acid sequence comprising a sequence that encodes an X chain element (XC element) of an dy heavy chain variable region (XCVR) and a Y chain element (YC element) of an antibody light chain variable region (YCVR), and wherein the XCVR and YCVR are matched, comprising: a) acquiring an isolated cell reaction site (e.g., an isolated cell reaction site described herein), e.g., a cell isolation micro-chamber, comprising: i) a cell (e.g., a cell described herein); and ii) a capture ate (e.g., a capture substrate described herein) capable of binding a first mRNA encoding an XCVR from the cell and a second mRNA encoding a YCVR from the cell; b) maintaining the isolated cell reaction site, e.g., the cell isolation micro-chamber, under conditions that allow lysis of the cell and g of the capture substrate with the first mRNA and the second mRNA to form the mRNA loaded capture substrate, wherein the isolated cell reaction site, e.g., cell isolation micro-chamber, does not e a nucleic acid encoding an XCVR or a YCVR from a cell other than the cell (e.g., a different cell); c) acquiring an isolated production reaction site (e.g., an isolated production reaction site described herein), e. g., a production micro-chamber, comprises: contacting the mRNA loaded capture substrate with a on mixture, e.g., a reaction mixture comprising reverse riptase, that uses the loaded mRNA as a template, to produce: a first double-stranded cDNA (ds cDNA) comprising a strand that is complementary to a first mRNA that encodes an XCVR from a cell; and a second ds cDNA comprising a strand complementary to a second mRNA ng a YCVR from the cell (the cDNA loaded e substrate); wherein the isolated production reaction site, e.g., a production chamber, does not include a nucleic acid encoding a YCVR or an XCVR from a cell other than the cell (e.g., a different cell). (1) maintaining the isolated production reaction site, e.g., the production micro-chamber, under conditions that allow amplification of the first and second ds cDNAs, to produce: a ity of XC ds cDNAs sing a segment that encodes an XC element of the XCVR from the cell, e.g., an XCVRS; and a plurality of YC ds cDNAs comprising a t that encodes a YC element of the YCVR from the cell, e.g., a YCVRS; e) acquiring an isolated linkage on site (e.g., an isolated linkage reaction site described ), e. g., a linkage micro-chamber, comprising: covalent linking, e. g., ligation, of a strand of the KC ds cDNA (XC strand) to a strand of the YC ds cDNA (YC strand), wherein the XC and YC strands are both sense strands or antisense strands; and f) amplifying the covalently linked, e.g., ligated, KC and YC strands.
In an embodiment, one or more (e.g., two, three, four, five, or all) of the steps a)-f) are performed in accordance with a method bed herein. In an embodiment, each of the steps a)-f) is performed in accordance with a method described herein.
In an aspect, the disclosure features a method of making a library comprising a plurality of unique members, the method comprising: making the plurality of members, wherein each of the members comprises a sequence that encodes an X chain element (XC element) of an X chain variable region (XCVR) and a Y chain element (YC element) of a Y chain variable region (YCVR), and wherein the XCVR and YCVR are matched, made by a method bed herein, n each unique nucleic acid sequence of the plurality comprises an XC element and a YC element from a different unique cell (e.g., a cell described herein), thereby making a library comprising a plurality of unique members.
In an embodiment, the plurality of unique members comprises at least 104, 105, 106, 107, 108, or 109 unique members. In an embodiment, the plurality of unique s comprises 104 to 109, 104 to 108, 104 to 107, 104 to 106, 104 to 105, 108 to 109, 107 to 109, 106 to 109, 105 to 109, 105 to 108, 106 to 107, 104 to 105, 105 to 106, 106 to 107, 107 to 108, or 108 to 109 unique members. In an embodiment, at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%, of the s in the library are unique members (which encode matched XC element and YC elements sequences). In an embodiment, less than 20%, 15%, 10%, 5%, 4%, 3%, 2%, or 1%, of the members in the library are unique members (which encode matched XC element and YC ts sequences).
In an aspect, the disclosure features a library comprising a plurality of unique members, wherein, i) each unique member of the plurality comprises a segment that encodes an XC element, e.g., an XCVRS, and a segment that encodes a YC element, e.g., a YCVRS, n the XC element and the YC element in each unique member is matched; ii) each unique member of the plurality comprises a segment that s an XC element, e.g., an XCVRS, and a segment that encodes a YC element, e.g., a YCVRS, from a ent unique cell; and iii) the library comprises one or more (e.g., two, three, four, or all) of the following ties: a) the library is made by a method described ; b) the plurality of unique members comprises at least 104, 105, 106, 107, 108, or 109 unique nucleic acid sequences; c) the plurality of unique members comprises 104 to 109, 104 to 108, 104 to 107, 104 to 106, 104 to 105, 108 to 109, 107 to 109, 106 to 109, 105 to 109, 105 to 108, 106 to 107, 104 to 105, 105 to 106, 106 to 107, 107 to 108, or 108 to 109 unique members; d) at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%, of the s in the library are unique members (which encode matched XC t and YC elements sequences); or e) less than 20%, 15%, 10%, 5%, 4%, 3%, 2%, or 1%, of the members in the library are unique members (which encode matched XC element and YC elements sequences).
In an embodiment, each unique member of the plurality is configured such that, when expressed, the XC element, e. g., the XCVRS, and the YC element, e.g., the YCVRS, form a functional antigen binding molecule, e.g., an scFV.
In an embodiment, the y is a display library. In an embodiment, each of the members of the plurality further encodes a polypeptide that results in display of the member on the surface of a display entity. In an embodiment, the library is a phage display library. In an ment, the y is a yeast y library. In an embodiment, the library is a mammalian display library.
In an aspect, the disclosure features a method of making a binding polypeptide (e.g., a polypeptide comprising an XC element and a YC element), the method comprising: a) acquiring a library described herein, e.g., by a method described herein; and b) expressing a polypeptide encoded by a unique nucleic acid of the library.
In an embodiment, the method further ses contacting the polypeptide with an n.
In an embodiment, the method further comprises retrieving (e.g., isolating or purifying) the nucleic acid that encodes a polypeptide that binds the n.
In an aspect, the disclosure features an isolated production reaction site, e.g., a production micro-chamber, which is an isolated tion reaction site described herein (e.g., comprising a nucleic acid encoding an XCVR and a nucleic acid encoding a YCVR, wherein the XCVR and the YCVR are matched).
In an embodiment, the ed production reaction site, e.g., a production micro-chamber, does not include a nucleic acid encoding an XCVR or a YCVR from a different cell.
In an ment, the isolated production reaction site, e.g., a production micro-chamber, comprises one, two, or all of: (i) one or more primers specific to V gene ces of the XC and YC; (ii) one or more s specific to overhangs introduced onto the XC and YC cDNAs; or (iii) one or more primers comprising a first member, a second member, and a third member comprising a nucleotide modification (e.g., a spacer) located between the first and second members, wherein the first member is capable of annealing with the second member of the same primer or a different primer, e.g., forming a structure comprising a duplex region of 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, l6, l7, l8, 19, 20, more basepairs.
In an embodiment, the isolated production reaction site, e.g., a tion micro-chamber, does not comprise a reagent that can covalently link nucleic acids, e.g., a ligase, e.g., a thermostable ligase. In another embodiment, the isolated production reaction site, e. g., a tion micro- chamber, comprises a reagent that can covalently link nucleic acids, e.g., a ligase, e.g., a thermostable ligase.
In an aspect, the disclosure features a self-annealing oligonucleotide comprising a first member, a second member, and third member comprising a nucleotide modification (e. g., a ) located between the first and second s, wherein the first member is capable of ing with the second member of the same oligonucleotide (e.g., for a method of making a nucleic acid sequence comprising a ce that encodes an XC element of an XCVR and a YC element of a YCVR, wherein the XCVR and YCVR are matched).
In an embodiment, the first and second members are capable of forming a hairpin structure comprising a duplex region of 4, 5,6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, more basepairs. In an embodiment, the first member is 5-40 nucleotides, e. 3., 5-10, 5-20, 5-30, 30-40, 20- 40, 10-30, 10-30, or 15-25 nucleotides, in length. In an embodiment, the second member is 5-40 nucleotides, e.g., 5-10, 5-20, 5-30, 30-40, 20-40, 10-30, 10-30, or 15-25 nucleotides, in length.
In an embodiment, the spacer is a spacer bed herein, e.g., a flexible spacer or a PEG spacer.
In an embodiment, the first member comprises a sequence that is complementary to the sequence of an oligonucleotide attached to a capture substrate.
In an embodiment, the second member comprises (e.g., from 5’ to 3’) one, two, or all of: (i) a ce that is complementary to at least a portion of the first member; (ii) a universal priming sequence (e.g., for PCR amplification or next-generation sequencing); and (iii) a sequence complementary to a target sequence, e.g., an XCVRS and/or a YCVRS. In an embodiment, the universal priming sequence is identical, or substantially identical, to the sequence that is complementary to at least a portion of the first member. In another embodiment, the universal g sequence is different from the sequence that is complementary to at least a portion of the first member. In an embodiment, the second member comprises a sequence for homologous recombination (e.g., in a yeast or mammalian cell).
In an , the disclosure features an isolated linkage reaction site, e. g., a linkage micro- chamber, which is an ed e reaction site described herein (e.g., sing a nucleic acid encoding an XCVR and a nucleic acid encoding a YCVR, n the XCVR and the YCVR are matched).
In an embodiment, the isolated linkage reaction site, e.g., a linkage micro-chamber, does not include a nucleic acid encoding an XCVR or a YCVR from a different cell.
In an ment, the ed linkage reaction site, e.g., a e micro-chamber, comprises a splint oligonucleotide (e.g., a splint ucleotide described herein) that is capable of hybridizing to a sequence comprising the junction of the XC strand and the YC strand, or a sequence mentary thereof, to form a duplexed region at the site of ligation.
In an embodiment, the isolated e reaction site, e.g., a linkage micro-chamber, comprises a reagent that can covalently link nucleic acids, e.g., a ligase, e.g., a thermostable ligase.
T-Cell Receptor Molecules The disclosures herein are not intended to be limited to antibody molecules. In an embodiment, the binding molecule is a TCR molecule, e. g., a soluble TCR molecule. In an embodiment, the binding molecule comprises a TCR or chain variable region and a TCR [3 chain variable . In an embodiment, the binding molecule comprises a TCR 7 chain variable region and a TCR 5 chain variable region. For example, in any of the aspects, embodiments, and definitions herein, an antibody heavy chain (or variable region) can be ed with a TCR or chain (or variable region), and an antibody light chain (or variable region) can be ed with a TCR [3 chain (or variable region); or an antibody heavy chain (or variable region) can be replaced with a TCR 7 chain (or variable region), and an antibody light chain (or variable region) can be replaced with a TCR 5 chain (or variable region).
Disclosed herein are T-cell receptor (TCR) molecules and libraries of TCR molecules. In an embodiment, the TCR molecule or y of TCR molecules are made by a method described herein.
As used herein, the term “TCR molecule,” also known as “T-cell receptor molecule” or “T cell receptor le,” refers to a protein, e.g., a TCR chain or a fragment thereof, comprising at least one TCR variable domain sequence. The term “TCR molecule” es, for example, full- , mature TCRs and antigen-binding nts of a TCR. For example, a TCR molecule can include an 0t chain variable domain sequence and a B chain variable domain sequence. In another example, a TCR molecule can include a y chain variable domain sequence and a 6 chain variable domain sequence. In an embodiment, the TCR molecule is a soluble TCR molecule.
T-cell receptors can be found on the surface of T cells and are responsible for recognizing fragments of antigen as peptides bound to major histocompatibility complex (MHC) molecules. A TCR lly e two different protein chains. In humans, in about 95% of T cells the TCR include an alpha (or) chain and a beta ([3) chain (encoded by TRA and TRB, respectively), whereas in about 5% of T cells the TCR include gamma and delta (7/5) chains (encoded by TRG and TRD, respectively). This ratio can change during ontogeny and in diseased states (such as leukemia).
When the TCR engages with antigenic peptide and MHC (peptide/MHC), the T lymphocyte is activated through signal transduction, e.g., a series of biochemical events mediated by associated enzymes, eptors, specialized r molecules, and activated or released transcription factors.
For example, the naturally-occurring TCR is typically a disulfide-linked ne-anchored heterodimeric protein including, e.g., the highly variable alpha (or) and beta ([3) chains expressed as part of a complex with the invariant CD3 chain molecules. T cells sing this receptor are sometimes referred to as azfi (or (XB) T cells, though a minority of T cells express an alternate or, formed by variable gamma (y) and delta (6) chains, sometimes referred as 1/5 T cells.
Each chain of TCR can include two extracellular domains: a variable (V) region and a constant (C) region. The constant region is proximal to the cell membrane, followed by a embrane region and a short cytoplasmic tail, while the variable region can bind to the peptide/MHC complex.
The variable domain of the TCR or chain or [5 chain each can have three hypervariable or mentarity determining regions (CDRs). There can also be an additional area of hypervariability on the [5 chain (HV4), which typically does not contact antigen and is not considered a CDR. The es in these variable domains are located in two regions of the TCR, at the interface of the or and [3 chains and in the [3-chain framework region that is in proximity to the CD3 signal- transduction complex. Without wishing to be bound by theory, it is believed that in an embodiment, CDR3 is the main CDR responsible for recognizing sed antigen. CDRl of the alpha chain can interact with the N—terminal part of the antigenic peptide, and CDRl of the [3 chain can interact with the C-terminal part of the peptide. CDR2 may recognize the MHC. CDR4 of the [3 chain is generally not thought to participate in n recognition, but may ct with ntigens.
The constant domain of the TCR include, e.g., short connecting sequences, which form a link between the two chains, e.g., h disulfide bonds.
The generation of TCR diversity arises mainly from genetic recombination of the DNA encoded segments in dual somatic T cells by somatic V(D)J recombination . Each recombined TCR may possess unique antigen specificity, determined by the ure of the antigen-binding site, e.g., formed by the 0t and [3 chains in case of (113 T cells or y and 5 chains on case of 1/5 T cells. For example, the TCR 0c and y chains can be generated by VJ recombination, and the [3 and 5 chains can be generated by VDJ recombination. The ection of these specific regions (e.g., V and J for 0t or 7 chain; V, D, and J for [3 or 5 chain) corresponds to the CDR3 region that is typically ant for peptide/MHC recognition.
The TCR receptor can form a complex of variable TCR chains (e.g., 0t and [3 chains with three dimeric signaling s CD35/e, CD3~yle and CD247 C/C or Cjn). T cell can express clonal TCRs which recognize specific peptide/MHC complex during physical contact between T cell and antigen- presenting cell-APC (MHC class II) or any other cell type (MHC class I). The signal from the T-cell complex can be enhanced by simultaneous binding of the MHC molecules by a ic co-receptor.
For example, on helper T cells and regulatory T cells, the co-receptor is CD4 that is specific for MHC class II, and on xic T cells, the co-receptor is CD8 that is specific for MHC class I.
The term “TCR” includes intact molecules as well as functional fragments thereof. TCR fragments may be obtained using any suitable method, ing several conventional techniques known to those with skill in the art, and the fragments can be screened for utility in the same manner as are intact TCRs. Constant s of the TCRs can be altered, e.g., mutated, to modify the properties of the TCR.
The TCR molecule can be a single chain TCR. The single chain TCR can be dimerized or multimerized to generate multivalent TCRs having icities for different epitopes of the same target protein.
The TCR molecules disclosed herein can also be single domain TCRs. Single domain TCRs can include TCRs whose complementary determining regions are part of a single domain polypeptide.
Examples include, but are not limited to, or, [3, y, or 5 chain TCRs, TCRs naturally devoid of a 0t, [3, y, or 5 chain, single domain TCRs derived from conventional two-chain TCRs, engineered TCRs and single domain scaffolds other than those derived from TCRs. Single domain TCRs may be any of the art, or any future single domain TCRs. Single domain TCRs may be derived from any species including, but not limited to mouse, human, camel, llama, fish, shark, goat, rabbit, and bovine.
The variable regions can be subdivided into regions of hypervariability, termed ementarity determining regions” (CDR), interspersed with regions that are more conserved, termed “framework regions” (FR or FW). The terms “complementarity determining ,” and “CDR,” as used herein in the context of TCR molecules refer to the sequences of amino acids within TCR variable s which confer antigen specificity and binding affinity. As used herein, the terms “framework,” “FW” and “FR” are used interchangeably.
As used herein, a “TCR variable domain sequence” refers to an amino acid sequence which can form the structure of a TCR variable domain. For e, the sequence may include all or part of the amino acid sequence of a naturally-occurring variable domain. For example, the sequence may or may not include one, two, or more N— or C-terminal amino acids, or may include other alterations that are compatible with ion of the protein structure.
The term “antigen-binding region” refers to the part of a TCR molecule that comprises determinants that form an interface that binds to an antigen, or an epitope thereof. With respect to proteins (or n mimetics), the antigen-binding region lly includes one or more loops (of at least, e.g., four amino acids or amino acid mimics) that form an ace that binds to the antigen.
Typically, the antigen-binding region of a TCR molecule includes at least one or two CDRs and/or hypervariable loops, or more typically at least three, four, five or six CDRs and/or hypervariable loops.
The terms “compete” or “cross-compete” are used interchangeably herein to refer to the ability of a TCR molecule to interfere with binding of another TCR molecule, to a . The interference with binding can be direct or indirect (e.g., h an allosteric modulation of the TCR molecule or the target). The extent to which an antibody molecule is able to interfere with the binding of another TCR molecule to the target, and therefore whether it can be said to compete, can be determined using a competition binding assay, for example, a FACS assay, an ELISA or BIACORE assay. In an embodiment, a competition binding assay is a quantitative ition assay. In an embodiment, a first TCR molecule is said to compete for g to the target with a second TCR le when the binding of the first antibody molecule to the target is reduced by 10% or more, e.g., 20% or more, 30% or more, 40% or more, 50% or more, 55% or more, 60% or more, 65% or more, 70% or more, 75% or more, 80% or more, 85% or more, 90% or more, 95% or more, 98% or more, 99% or more in a competition binding assay (e.g., a competition assay described herein).
The TCR molecule can be a polyclonal or a monoclonal. In some embodiments, the TCR can be recombinantly produced, e.g., produced by any suitable phage display or combinatorial methods.
Phage y and combinatorial methods are known in the art.
In an embodiment, the TCR molecule is a fully human TCR (e. g., a TCR made in a mouse which has been genetically engineered to produce a TCR from a human TCR sequence), or a non- human TCR, e.g., a rodent (mouse or rat), goat, e (e.g., monkey), camel TCR. In an embodiment, the non-human TCR is a rodent (mouse or rat TCR). For example, Human TCRs can be generated using transgenic mice carrying the human TCR genes rather than the mouse system.
A TCR can be one in which the variable region, or a portion thereof, e.g., the CDRs, are generated in a non-human organism, e.g., a rat or mouse. Chimeric, CDR-grafted, and humanized TCRs are within the invention. TCRs generated in a non-human sm, e.g., a rat or mouse, and then modified, e.g., in the variable framework or constant region, to decrease antigenicity in a human are within the invention.
Chimeric TCRs can be produced by any suitable recombinant DNA technique.
A humanized or CDR-grafted TCR will have at least one or two but generally all three recipient CDRs (of TCR chains) replaced with a donor CDR. The TCR may be replaced with at least a portion of a non-human CDR or only some of the CDRs may be replaced with non-human CDRs. It is only necessary to replace the number of CDRs required for g of the humanized TCR to an antigen. In an embodiment, the donor will be a rodent TCR, e.g., a rat or mouse TCR, and the ent will be a human framework or a human consensus framework. Typically, the TCR providing the CDRs is called the “donor” and the TCR providing the ork is called the tor.” In some embodiments, the donor TCR is a non-human (e.g., rodent). The acceptor framework is lly a naturally-occurring (e.g., a human) framework or a sus framework, or a sequence about 85% or higher, e.g., 90%, 95%, 99% or higher identical thereto.
As used herein, the term “consensus sequence” refers to the ce formed from the most frequently occurring amino acids (or nucleotides) in a family of related sequences (See e.g., Winnaker, From Genes to Clones (Verlagsgesellschaft, Weinheim, Germany 1987). In a family of proteins, each position in the sus sequence is ed by the amino acid occurring most frequently at that position in the family. If two amino acids occur equally frequently, either can be included in the consensus sequence. A “consensus framework” refers to the framework region in the consensus TCR sequence.
A TCR can be humanized by any suitable method. Humanized or CDR-grafted TCR s can be produced by CDR-grafting or CDR substitution, wherein one, two, or all CDRs of an immunoglobulin chain can be replaced. Also provided are humanized TCRs in which specific amino acids have been substituted, deleted or added.
In an embodiment, the TCR molecule has a nt region. The constant region can be d, e.g., mutated, to modify a property of the TCR molecule. In an embodiment, a constant region of the TCR molecule is altered. Methods for altering a constant region are known in the art.
In an embodiment, the only amino acids in the TCR molecule are cal amino acids. In an ment, the TCR molecule comprises naturally-occurring amino acids; analogs, derivatives and congeners thereof; amino acid analogs having variant side chains; and/or all stereoisomers of any of any of the ing. The TCR molecule may comprise the D- or L— optical isomers of amino acids and peptidomimetics.
A polypeptide of a TCR le described herein may be linear or branched, it may comprise modified amino acids, and it may be interrupted by non-amino acids. The TCR molecule may also be modified; for example, by disulfide bond formation, glycosylation, lipidation, acetylation, phosphorylation, or any other manipulation, such as conjugation with a labeling component. The polypeptide can be isolated from natural sources, can be a produced by recombinant techniques from a otic or prokaryotic host, or can be a product of synthetic ures.
The TCR molecule described herein can be used alone in unconjugated form, or can be bound to a substance, e.g., a toxin or moiety (e.g., a therapeutic drug; a compound emitting radiation; molecules of plant, fungal, or bacterial origin; or a biological protein (e. g., a protein toxin) or le (e. g., a recombinant viral particle, e.g., via a viral coat n). For example, the TCR molecule can be coupled to a radioactive isotope such as an (1-, [3-, or y—emitter, or a B-and y—emitter.
A TCR molecule can be derivatized or linked to r functional molecule (e.g., another peptide or protein). As used herein, a “derivatized” TCR le is one that has been modified.
Methods of derivatization include but are not limited to the addition of a fluorescent moiety, a ucleotide, a toxin, an enzyme or an affinity ligand such as biotin. Accordingly, the TCR molecules are intended to include derivatized and otherwise modified forms of the antibodies described herein, including immunoadhesion molecules. For example, a TCR molecule can be functionally linked (by chemical coupling, c fusion, noncovalent association or otherwise) to one or more other lar entities, such as another TCR (e.g., a bispecific TCR), a detectable agent, a toxin, a pharmaceutical agent, and/or a protein or peptide that can mediate association of the TCR or TCR portion with another molecule (such as a streptavidin core region or a polyhistidine tag).
Some types of derivatized TCR le are produced by inking two or more TCRs (of the same type or of different types, e.g., to create bispecific TCRs). Suitable crosslinkers include those that are heterobifunctional, having two distinctly reactive groups separated by an appropriate spacer (e.g., m—maleimidobenzoyl-N-hydroxysuccinimide ester) or homobifunctional (e.g., disuccinimidyl suberate). Such linkers are available from Pierce Chemical Company, Rockford, Ill.
Useful able agents with which a TCR molecule may be derivatized (or d) to include fluorescent compounds, various enzymes, prosthetic groups, luminescent materials, bioluminescent materials, fluorescent emitting metal atoms, e.g., um (Eu), and other anthanides, and radioactive materials (described below). Exemplary cent detectable agents include fluorescein, cein isothiocyanate, rhodamine, 5dimethylamine-l-napthalenesulfonyl chloride, phycoerythrin and the like. A TCR may also be derivatized with detectable enzymes, such as alkaline phosphatase, horseradish peroxidase, B-galactosidase, acetylcholinesterase, glucose oxidase and the like. When a TCR is derivatized with a detectable enzyme, it is detected by adding additional reagents that the enzyme uses to produce a able reaction product. For e, when the detectable agent horseradish peroxidase is present, the addition of hydrogen peroxide and diaminobenzidine leads to a colored reaction product, which is detectable. A TCR molecule may also be tized with a prosthetic group (e.g., streptavidin/biotin and avidin/biotin). For example, a TCR may be derivatized with biotin, and detected through indirect measurement of avidin or streptavidin binding.
Examples of suitable fluorescent materials include umbelliferone, cein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin; an example of a luminescent al includes luminol; and es of bioluminescent materials include luciferase, luciferin, and aequorin.
Labeled TCR molecules can be used, for example, diagnostically and/or experimentally in a number of contexts, including (i) to isolate a predetermined antigen by standard techniques, such as ty chromatography or immunoprecipitation; (ii) to detect a predetermined antigen (e.g., in a cellular lysate or cell supernatant) in order to evaluate the abundance and pattern of expression of the protein; (iii) to monitor protein levels in tissue as part of a clinical testing procedure, e.g., to determine the efficacy of a given treatment regimen.
A TCR molecule may be ated to another molecular entity, typically a label or a therapeutic (e.g., antimicrobial (e.g., antibacterial or bactericidal), immunomodulatory, immunostimularoty, cytotoxic, or cytostatic) agent or moiety. Radioactive isotopes can be used in stic or therapeutic applications. Radioactive isotopes that can be coupled to the antibody molecules include, but are not limited to (1-, [3-, or y—emitters, or B-and y—emitters. Such ctive isotopes include, but are not limited to iodine (1311 or I), yttrium (90Y), lutetium ), actinium (225Ac), praseodymium, astatine , rhenium (186Re), bismuth (212Bi or 213Bi), indium (1 1 1In), technetium (99mTc), orus (32F), rhodium (lgth), sulfur (35$) carbon (14C), tritium (3H), chromium (SlCr), chlorine (36Cl), cobalt (“Co or 58C0), iron (59Fe), selenium (7586), or gallium (67Ga).
Radioisotopes useful as therapeutic agents include yttrium (90Y), lutetium (177Lu), actinium (225Ac), praseodymium, astatine (leAt), rhenium ), bismuth (212Bi or 213Bi), and rhodium (lgth).
Radioisotopes useful as labels, e.g., for use in diagnostics, include iodine (1311 or 125I), indium (mln), technetium (99mTc), phosphorus (32F), carbon (14C), and tritium (3H), or one or more of the eutic isotopes listed above.
The present sure provides radiolabeled TCR molecules and s of labeling the same. In an embodiment, a method of ng a TCR molecule is disclosed. The method includes contacting a TCR molecule, with a chelating agent, to thereby produce a ated TCR. The conjugated antibody is radiolabeled with a radioisotope, e.g., 111Indium, ium and 177Lutetium, to thereby produce a labeled TCR molecule.
In some aspects, this disclosure es a method of making a TCR molecule disclosed herein. The method includes: providing an antigen, or a fragment thereof; obtaining a TCR molecule that specifically binds to the n; evaluating efficacy of the TCR molecule in ting activity of the antigen and/or organism sing the antigen. The method can further include administering the TCR molecule, including a derivative thereof (e.g., a humanized TCR molecule) to a subject, e.g., a human.
This disclosure provides an isolated c acid molecule encoding the above TCR molecule, vectors and host cells thereof. The nucleic acid molecule includes, but is not limited to, RNA, c DNA and cDNA.
Animal Models The polypeptides (e.g., binding polypeptides, e.g., dy molecules or TCR molecules) described herein can be evaluated in viva, e.g., using various animal models. For example, an animal model can be used to test the efficacy of a binding polypeptide (e.g., an antibody molecule or TCR molecule) bed herein in modulating a biological on of a target molecule or cell. As another example, an animal model can also be used to test the efficacy of a binding polypeptide (e.g., an antibody molecule or TCR molecule) described herein in in treating, preventing, or diagnosing a disorder described . Animal models can also be used, e.g., to igate for side effects, measure concentrations of binding ptides (e.g., antibody molecules or TCR molecules) in situ, demonstrate correlations between a function of a target molecule or cell and a disorder described herein.
Exemplary animal models for other disorders described herein are also known in the art.
Exemplary types of animals that can be used to evaluate the binding polypeptides (e.g., antibody molecules or TCR molecules) bed herein include, but are not limited to, mice, rats, rabbits, guinea pigs, and monkeys.
Pharmaceutical Compositions and Kits In some aspects, this disclosure provides compositions, e.g., pharmaceutically acceptable compositions, which include a polypeptide (e.g., a binding polypeptide, e.g., an antibody molecule or a TCR molecule) described herein, ated together with a ceutically acceptable carrier.
As used herein, “pharmaceutically acceptable carrier” includes any and all ts, dispersion media, isotonic and absorption delaying agents, and the like that are physiologically compatible. The r can be suitable for intravenous, intramuscular, subcutaneous, parenteral, rectal, spinal or mal administration (e.g., by ion or infusion). In certain embodiments, less than about 5%, e.g., less than about 4%, 3%, 2%, or 1% of the binding polypeptides in the pharmaceutical composition are present as aggregates. In other embodiments, at least about 95%, e.g., at least about 96%, 97%, 98%, 98.5%, 99%, 99.5%, 99.8%, or more of the binding polypeptides in the pharmaceutical composition are present as monomers. In some embodiments, the level of aggregates or monomers is ined by chromatography, e.g., high performance size exclusion chromatography (HP-SEC).
The compositions set out herein may be in a variety of forms. These include, for example, liquid, semi-solid and solid dosage forms, such as liquid solutions (e.g., injectable and infusible solutions), dispersions or suspensions, liposomes, and suppositories. A le form depends on the intended mode of administration and therapeutic application. Typical suitable compositions are in the form of injectable or infusible ons. One suitable mode of stration is parenteral (e.g., intravenous, aneous, intraperitoneal, intramuscular). In an embodiment, the binding polypeptide is administered by intravenous infusion or injection. In another embodiment, the binding polypeptide is administered by intramuscular or subcutaneous injection.
The phrases “parenteral administration” and “administered parenterally” as used herein means modes of administration other than enteral and topical stration, usually by injection, and includes, without limitation, intravenous, intramuscular, rterial, intrathecal, apsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal, epidural and intrasternal injection and infusion.
Therapeutic compositions typically should be sterile and stable under the ions of manufacture and storage. The composition can be ated as a solution, microemulsion, dispersion, liposome, or other ordered structure suitable to high concentrations of binding polypeptides (e.g., dy molecules or TCR molecules). Sterile injectable solutions can be prepared by incorporating the active compound (e.g., binding polypeptide) in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the active compound into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable ons, the preferred methods of preparation are vacuum drying and freeze-drying that yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof. The proper fluidity of a solution can be maintained, for example, by the use of a coating such as in, by the maintenance of the ed particle size in the case of dispersion and by the use of surfactants. ged absorption of injectable compositions can be brought about by including in the composition an agent that delays absorption, for example, monostearate salts and gelatin.
The binding polypeptides (e.g., antibody molecules or TCR ors) described herein can be administered by a variety of methods. Several are known in the art, and for many therapeutic, prophylactic, or stic applications, an appropriate mode of administration is intravenous injection or infusion. For example, the binding polypeptides can be administered by intravenous infusion at a rate of less than lOmg/min; preferably less than or equal to 5 mg/min to reach a dose of about 1 to 100 mg/mz, preferably about 5 to 50 mg/m2, about 7 to 25 mg/m2 and more preferably, about 10 mg/m2. As will be appreciated by the skilled artisan, the route and/or mode of administration will vary ing upon the d results. In certain embodiments, the active compound may be prepared with a carrier that will t the nd against rapid release, such as a controlled release formulation, including implants, transdermal patches, and microencapsulated delivery systems. Biodegradable, biocompatible rs can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Many methods for the preparation of such formulations are patented or generally known to those skilled in the art. See, e.g., Sustained and Controlled Release Drug Delivery Systems, J. R. Robinson, ed., Marcel Dekker, Inc., New York, 1978.
In certain embodiments, the binding polypeptide (e. g., antibody molecule or TCR molecule) can be orally administered, for example, with an inert diluent or an assimilable edible carrier. The binding polypeptide (and other ients, if desired) may also be enclosed in a hard or soft shell gelatin capsule, compressed into tablets, or orated directly into the subject’s diet. For oral therapeutic administration, the binding polypeptide may be incorporated with excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, , , and the like. To administer the binding polypeptide (e. g., dy molecule) by other than parenteral administration, it may be necessary to coat the compound with, or co-administer the nd with, a material to prevent its inactivation. Therapeutic, prophylactic, or diagnostic compositions can also be administered with medical devices, and several are known in the art.
Dosage regimens are adjusted to provide the d response (e. g., a eutic, lactic, or diagnostic response). For e, a single bolus may be administered, several divided doses may be administered over time or the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation. It is especially advantageous to formulate parenteral compositions in dosage unit form for ease of administration and mity of dosage.
Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the subjects to be treated; each unit contains a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The specification for the dosage unit forms are dictated by and directly dependent on (a) the unique characteristics of the antibody molecule and the particular therapeutic, prophylactic, or stic effect to be achieved, and (b) the limitations inherent in the art of compounding such a binding polypeptide for the treatment of sensitivity in individuals.
An exemplary, non-limiting range for a therapeutically, prophylactically, or diagnostically effective amount of a binding ptide (e. g., an antibody le or TCR molecule) is about 0.1- 50 mg/kg, e.g., about 0.1-30 mg/kg, e.g., about 1-30, 1-15, 1-10, 1-5, 5-10, or 1-3 mg/kg, e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 30, 40, or 50 mgikg. The binding polypeptide can be administered by intravenous infusion at a rate of less than 10 mg/min, e. g., less than or equal to 5 mg/min to reach a dose of about 1 to 100 mg/mz, e.g., about 5 to 50 mg/m2, about 7 to 25 mg/m2, e.g., about 10 mg/m2.
It is to be noted that dosage values may vary with the type and severity of the condition to be ated. It is to be further understood that for any particular t, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the compositions, and that dosage ranges set forth herein are exemplary only and are not ed to limit the scope or practice of the claimed compositions.
The pharmaceutical compositions herein may include a “therapeutically effective amount,” “prophylactically effective amount,” or “diagnostically effectively amount” of a binding polypeptide (e. g., an dy molecule or TCR molecule) described herein.
A “therapeutically effective amount” refers to an amount effective, at s and for periods of time necessary, to e the desired therapeutic result. A therapeutically effective amount of the binding polypeptide (e.g., antibody molecule or TCR molecule) may vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of the antibody or antibody n to elicit a desired response in the individual. A therapeutically ive amount is also one in which any toxic or ental effect of the antibody molecule is outweighed by the therapeutically beneficial effects.
A “therapeutically effective dosage” typically inhibits a measurable parameter by at least about 20%, e.g., by at least about 40%, by at least about 60%, or by at least about 80% relative to untreated subjects. The measurable ter may be, e.g., hematuria, colored urine, foamy urine, pain, swelling (edema) in the hands and feet, or high blood pressure. The ability of a binding polypeptide (e.g., an dy molecule) to inhibit a able parameter can be evaluated in an animal model system predictive of efficacy in treating or preventing a disorder described herein.
Alternatively, this property of a composition can be evaluated by examining the ability of the binding polypeptide (e.g., antibody molecule or TCR molecule) to modulate a biological function of a target molecule or cell, e.g., by an in vitro assay.
A “prophylactically effective amount” refers to an amount effective, at s and for periods of time necessary, to achieve the desired prophylactic result. Typically, since a prophylactic dose is used in subjects prior to or at an earlier stage of disease, the prophylactically effective amount will be less than the therapeutically effective amount.
A “diagnostically effective amount” refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired diagnostic result. Typically, a diagnostically effective amount is one in which a disorder, e.g., a er described herein, can be diagnosed in vitro, ex vivo, or in viva.
Also within this sure is a kit that comprises a binding polypeptide (e. g., an antibody molecule or TCR molecule), described herein. The kit can include one or more other elements including: instructions for use; other ts, e.g., a label, a therapeutic agent, or an agent useful for chelating, or otherwise coupling, an dy molecule to a label or eutic agent, or a radioprotective composition; devices or other materials for preparing the binding polypeptide (e.g., antibody molecule or TCR molecule) for administration; pharmaceutically acceptable carriers; and devices or other materials for administration to a subject.
Nucleic Acids The t disclosure also features nucleic acids sing nucleotide sequences that encode polypeptides (e. g., binding polypeptides, e.g., dy molecules or T cell receptor molecules), as described herein.
In an embodiment, the c acid further comprises a nucleotide sequence ng a heavy chain variable region of a polypeptide (6.3., an antibody molecule or TCR molecule) bed herein, or having a tide sequence substantially homologous thereto (e.g., a sequence at least about 85%, 90%, 95%, 99% or more identical thereto, and/or e of hybridizing under the stringency conditions described herein). In another embodiment, the nucleic acid further comprises a nucleotide sequence encoding a light chain variable region of a polypeptide (e.g., an antibody molecule or TCR molecule) described herein, or a nucleotide sequence substantially homologous thereto (e.g., a sequence at least about 85%, 90%, 95%, 99% or more identical thereto, and/or capable of izing under the stringency conditions described herein). In yet another embodiment, the nucleic acid further comprises a tide sequence encoding a heavy chain variable region and a light chain variable region of a polypeptide (e.g., an antibody molecule or TCR molecule) described herein, or a nucleotide sequence substantially homologous thereto (e.g., a sequence at least about 85%, 90%, 95%, 99% or more identical thereto, and/or capable of hybridizing under the stringency ions described herein).
In an embodiment, the nucleic acid further comprises a nucleotide sequence encoding at least one, two, or three CDRs from a heavy chain variable region of a polypeptide (e.g., an antibody molecule or TCR molecule) bed herein, or a nucleotide sequence substantially homologous thereto (e.g., a sequence at least about 85%, 90%, 95%, 99% or more identical thereto, and/or capable of hybridizing under the stringency conditions described herein). In another embodiment, the nucleic acid further comprises a nucleotide sequence encoding at least one, two, or three CDRs from a light chain variable region of a polypeptide (e.g., an antibody le or TCR molecule) described herein, or a nucleotide sequence substantially homologous thereto (e.g., a sequence at least about 85%, 90%, 95%, 99% or more identical thereto, and/or capable of hybridizing under the ency conditions described herein). In yet another embodiment, the nucleic acid comprises a tide sequence encoding at least one, two, three, four, five, or six CDRs from heavy and light chain variable regions of a ptide (e.g., an antibody le or TCR le) described herein, or a nucleotide sequence substantially homologous thereto (e.g., a sequence at least about 85%, 90%, 95%, 99% or more identical thereto, and/or capable of hybridizing under the stringency conditions described herein) .
In an embodiment, the nucleic acid comprises a portion of a nucleotide sequence described herein. The portion may encode, for example, a variable region (e.g., VH or VL); one, two, or three or more (e.g., four, five, or six) CDRs; or one, two, three, or four or more framework regions, optionally, a constant region or an Fc region.
The c acids disclosed herein include deoxyribonucleotides or cleotides, or analogs thereof. The polynucleotide may be either single-stranded or double-stranded, and if - ed may be the coding strand or non-coding (antisense) strand. A polynucleotide may comprise modified nucleotides, such as methylated tides and nucleotide analogs. The sequence of nucleotides may be interrupted by non-nucleotide components. A polynucleotide may be further modified after polymerization, such as by conjugation with a labeling component. The nucleic acid may be a recombinant polynucleotide, or a cleotide of genomic, cDNA, semisynthetic, or synthetic origin which either does not occur in nature or is linked to another polynucleotide in a non- natural ement.
In some aspects, the ation features host cells and vectors containing the nucleic acids described herein. The nucleic acids may be present in a single vector or separate vectors present in the same host cell or separate host cell, as described in more detail below.
Vectors The present disclosure features s that comprise nucleotide sequences encoding polypeptides (e.g., binding polypeptides, e.g., antibody molecules or TCR molecules).
The vectors include, but are not limited to, a virus, plasmid, cosmid, lambda phage or a yeast artificial some (YAC).
Numerous vector systems can be employed. For example, one class of vectors utilizes DNA elements which are d from animal viruses such as, for example, bovine papilloma virus, polyoma Virus, adenovirus, ia virus, baculovirus, retroviruses (Rous Sarcoma Virus, MMTV or MOMLV) or SV4O virus. Another class of vectors utilizes RNA elements derived from RNA viruses such as Semliki Forest Virus, Eastern Equine Encephalitis Virus and Flaviviruses.
Additionally, cells which have stably integrated the DNA into their chromosomes may be selected by introducing one or more markers which allow for the selection of transfected host cells.
The marker may provide, for example, prototropy to an auxotrophic host, biocide resistance (e.g., antibiotics), or resistance to heavy metals such as copper, or the like. The able marker gene can be either directly linked to the DNA sequences to be expressed, or introduced into the same cell by sformation. Additional elements may also be needed for l synthesis of mRNA. These elements may e splice signals, as well as transcriptional promoters, enhancers, and termination signals.
Once the expression vector or DNA sequence containing the constructs has been prepared for expression, the expression vectors may be transfected or introduced into an appropriate host cell.
Various techniques may be ed to achieve this, such as, for example, protoplast fusion, calcium phosphate precipitation, oporation, retroviral transduction, viral ection, gene gun, lipid based transfection or other conventional techniques. In the case of protoplast fusion, the cells are grown in media and screened for the appropriate activity.
Methods and conditions for culturing the resulting transfected cells and for recovering the polypeptide (e. g., antibody molecule) produced are known to those skilled in the art, and may be varied or optimized depending upon the specific expression vector and mammalian host cell employed, based upon the present description.
Cells The present disclosure also provides host cells comprising a nucleic acid encoding a polypeptide (e.g., an antibody le or TCR le) as described herein. The ptide (e.g., antibody molecule or TCR molecule) can be engineered in accordance with a method described herein. For example, the host cells may se a nucleic acid molecule having a nucleotide sequence of a polypeptide described herein (e.g., an antibody molecule or TCR molecule bed herein), a sequence substantially homologous thereto (e.g., a sequence at least about 85%, 90%, 95%, 99% or more identical o, and/or capable of hybridizing under the stringency conditions described ), or a portion of one of said nucleic acids.
In some embodiments, the host cells are genetically ered to comprise nucleic acids encoding the polypeptide (e.g., antibody molecule or TCR molecule) described herein.
In certain embodiments, the host cells are genetically engineered by using an expression cassette. The phrase “expression cassette,” refers to nucleotide sequences, which are capable of affecting expression of a gene in hosts compatible with such sequences. Such cassettes may include a promoter, an open reading frame with or without introns, and a termination signal. Additional factors necessary or helpful in ing expression may also be used, such as, for example, an inducible promoter.
The disclosure also provides host cells comprising the vectors described herein.
The cell can be, but is not d to, a eukaryotic cell, a bacterial cell, an insect cell, or a human cell. Suitable eukaryotic cells include, but are not limited to, Vero cells, HeLa cells, COS cells, CHO cells, HEK293 cells, BHK cells and MDCKII cells. Suitable insect cells include, but are not limited to, Sf9 cells.
Uses of Polypeptides The polypeptides (e.g., g polypeptides, e.g., antibody molecules or TCR molecule) disclosed herein, as well as the pharmaceutical compositions sed herein, have in vitro, ex vivo, and in viva therapeutic, prophylactic, and/or diagnostic utilities.
In an embodiment, the polypeptide (e.g., antibody molecule or TCR molecule) modulates (e.g., s (e.g., ts, blocks, or neutralizes) or increases (e.g., activates, initiates, or es)) one or more biological activities of a target molecule (e. g., protein) or cell. For example, these polypeptides (e.g., antibody les or TCR molecules) can be administered to cells in culture, in vitra or ex viva, or to a subject, e.g., a human subject, e.g., in viva, to modulate one or more biological activities of the target molecule or cell. Accordingly, in an aspect, the disclosure provides a method of treating, preventing, or diagnosing a disorder, e. g., a er described herein, in a subject, comprising administering to the t a polypeptide (e.g., an antibody molecule or TCR molecule) described herein, such that the disorder is treated, prevented, or diagnosed. For example, the disclosure provides a method comprising contacting the polypeptide (e.g., antibody le or TCR molecule) described herein with cells in e, 6.3. in vitra or ex viva, or administering the polypeptide (e.g., antibody molecule or TCR molecule) described herein to a subject, e.g., in viva, to treat, prevent, or diagnose a disorder, e.g., a disorder associated with a target molecule or cell (e.g., a disorder described ).
As used herein, the term “subject” is intended to include human and non-human animals. In some embodiments, the t is a human t, e.g., a human patient having a disorder described herein, or at risk of having a disorder described herein. The term uman animals” includes mammals and non-mammals, such as non-human primates. In an embodiment, the subject is a human. The methods and compositions described herein are suitable for treating human patients for a disorder described herein. Patients having a disorder described herein include those who have developed a disorder described herein but are (at least temporarily) asymptomatic, patients who have exhibited a symptom of a disorder described herein, or patients having a disorder related to or associated with a disorder described herein.
Methods of Treating or Preventing Disorders The polypeptides (e.g., antibody molecules or TCR molecules) described herein can be used to treat or prevent disorders or conditions. In an embodiment, the polypeptide has an optimal or improved half-life, which can be desirable for treating or ting the disorder or condition. While not wishing to be bound by , it is believed that in an embodiment, the ptide described herein (e.g., the polypeptide having an optimal or improved half-life) can provide one or more benefits over another polypeptide having the same or similar g affinity and/or icity (e.g., a polypeptide that does not have, or has not been engineered to have, an l or improved half-life).
These benefits can include, but are not limited to, an increased therapeutic or preventive efficacy, a d dosage regimen, or an improved pharmacokinetic ty. In an embodiment, the polypeptide includes a mutated Fc region as described herein.
Exemplary disorders or conditions that can be treated or ted by the polypeptides described herein include, but are not limited to, a cancer (e.g., a solid tumor or a hematologic cancer), an infectious disease (e.g., a bacterial infection or a viral infection), an immune disorder (e.g., an autoimmune er), an inflammatory disorder, a metabolic disorder (e.g., es), a cardiovascular er, an organ transplant rejection.
Exemplary cancers that can be d or prevented by the polypeptides described herein include, but are not limited to, acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), adrenocortical carcinoma, Kaposi sarcoma, an AIDS -related lymphoma, y central nervous system (CNS) lymphoma, anal cancer, appendix cancer, astrocytoma, atypical teratoid/rhabdoid tumor, basal cell carcinoma, bile duct cancer, bladder cancer, bone cancer (e.g., Ewing sarcoma or osteosarcoma and malignant fibrous histiocytoma), brain tumor (e.g., astrocytomas, brain stem glioma, central nervous system atypical teratoid/rhabdoid tumor, central nervous system embryonal tumor, central nervous system germ cell tumor, craniopharyngioma, or ependymoma), breast cancer, bronchial tumor, Burkitt lymphoma, carcinoid tumor (e.g., gastrointestinal carcinoid tumor), cardiac (heart) tumor, embryonal tumor, germ cell tumor, lymphoma, cervical cancer, cholangiocarcinoma, ma, chronic lymphocytic leukemia (CLL), chronic myelogenous ia (CML), chronic myeloproliferative neoplasm, colon cancer, colorectal cancer, craniopharyngioma, ous T-cell lymphoma, ductal carcinoma in situ (DCIS), endometrial cancer, ependymoma, esophageal cancer, esthesioneuroblastoma, Ewing sarcoma, extracranial germ cell tumor, extragonadal germ cell tumor, eye cancer (e.g., intraocular melanoma or retinoblastoma), ian tube cancer, fibrous histiocytoma of bone, osteosarcoma, gallbladder cancer, gastric (stomach) cancer, gastrointestinal oid tumor, gastrointestinal stromal tumors (GIST), germ cell tumor (e. g., central nervous system tumor, extracranial tumor, extragonadal tumor, ovarian cancer, or ular cancer), gestational trophoblastic disease, glioma, hairy cell leukemia, head and neck cancer, hepatocellular (liver) cancer, Hodgkin lymphoma, hypopharyngeal cancer, cular melanoma, islet cell tumor, pancreatic neuroendocrine tumor, Kaposi sarcoma, kidney cancer (e.g., renal cell cancer or Wilms tumor), Langerhans cell histiocytosis (LCH), laryngeal cancer, leukemia (e.g., acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), chronic lymphocytic ia (CLL), chronic myelogenous leukemia (CML), or hairy cell ia), lip and oral cavity cancer, liver cancer, lung cancer (e.g., all cell lung cancer (NSCLC) or small cell lung cancer), ma (e.g., elated, Burkitt lymphoma, ous T-cell lymphoma, Hodgkin lymphoma, non-Hodgkin lymphoma, or primary central nervous system (CNS) lymphoma), Waldenstrom lobulinemia, male breast cancer, malignant fibrous histiocytoma of bone and osteosarcoma, melanoma (e.g., intraocular (eye) melanoma), Merkel cell carcinoma, mesothelioma, metastatic squamous neck cancer, midline tract carcinoma, mouth cancer, le endocrine neoplasia syndrome, multiple myeloma/plasma cell neoplasm, mycosis fungoides, myelodysplastic me, myelodysplastic/myeloproliferative neoplasm, chronic myeloproliferative sm, nasal cavity and paranasal sinus cancer, nasopharyngeal cancer, neuroblastoma, oral cancer, lip and oral cavity cancer, oropharyngeal cancer, osteosarcoma and malignant fibrous histiocytoma of bone, n cancer (e.g., epithelial ovarian cancer or germ cell ovarian tumor), pancreatic cancer, atic neuroendocrine tumors (islet cell tumors), papillomatosis, paraganglioma, paranasal sinus and nasal cavity cancer, parathyroid cancer, penile cancer, pharyngeal cancer, pheochromocytoma, pituitary tumor, pleuropulmonary blastoma, neal cancer, prostate cancer, rectal cancer, retinoblastoma, rhabdomyosarcoma, salivary gland cancer, sarcoma (e. g., Ewing sarcoma, Kaposi sarcoma, osteosarcoma, myosarcoma, soft tissue sarcoma, or uterine sarcoma), Sézary syndrome, skin cancer (e.g., melanoma, Merkel cell carcinoma, or nonmelanoma skin cancer), small intestine cancer, us cell carcinoma, testicular cancer, throat cancer, thymoma and thymic carcinoma, thyroid cancer, transitional cell cancer of the renal pelvis and ureter, urethral cancer, endometrial uterine cancer, vaginal cancer, vulvar , or a metastatic lesion thereof. ary infectious diseases that can be treated or prevented by the polypeptides described herein include, but are not limited to, Acinetobacter infections, actinomycosis, African sleeping sickness (African trypanosomiasis), AIDS (acquired immunodeficiency syndrome), amebiasis, anaplasmosis, angiostrongyliasis, anisakiasis, anthrax, arcanobacterium haemolyticum infection, argentine hemorrhagic fever, asis, aspergillosis, astrovirus infection, babesiosis, bacillus cereus infection, bacterial pneumonia, bacterial vaginosis, bacteroides ion, balantidiasis, bartonellosis, baylisascaris ion, bk virus infection, black , blastocystosis, mycosis, bolivian hemorrhagic fever, botulism (and infant botulism), brazilian hemorrhagic fever, brucellosis, bubonic plague, burkholderia infection, buruli ulcer, calicivirus ion (norovirus and sapovirus), campylobacteriosis, candidiasis (moniliasis; thrush), ariasis, carrion’s disease, cat-scratch disease, itis, chagas disease (american trypanosomiasis), chancroid, chickenpox, chikungunya, chlamydia, chlamydophila pneumoniae ion (taiwan acute respiratory agent or twar), cholera, chromoblastomycosis, chytridiomycosis, clonorchiasis, clostridium difficile colitis, coccidioidomycosis, colorado tick fever (CTF), common cold (Acute viral rhinopharyngitis; Acute coryza), Creutzfeldt-Jakob disease (CJD), Crimean-Congo hemorrhagic fever (CCHF), cryptococcosis, cryptosporidiosis, cutaneous larva migrans (CLM), cyclosporiasis, cysticercosis, cytomegalovirus infection, dengue fever, desmodesmus ion, dientamoebiasis, diphtheria, diphyllobothriasis, dracunculiasis, ebola hemorrhagic fever, echinococcosis, hiosis, enterobiasis rm infection), coccus infection, enterovirus infection, epidemic typhus, erythema infectiosum (fifth disease), em subitum (sixth disease), fasciolasis, fasciolopsiasis, fatal familial insomnia (FFI), filariasis, food poisoning by clostridium perfringens, free-living amebic infection, fusobacterium ion, gas gangrene (clostridial myonecrosis), geotrichosis, gerstmann- straussler-scheinker syndrome (GSS), giardiasis, glanders, gnathostomiasis, gonorrhea, granuloma inguinale (donovanosis), Group A streptococcal infection, Group B streptococcal infection, haemophilus influenzae infection, hand, foot and mouth disease (HFMD), Hantavirus ary me (HPS), heartland virus disease, helicobacter pylori infection, hemolytic-uremic syndrome (HUS), hemorrhagic fever with renal me (HFRS), hepatitis A, hepatitis B, hepatitis C, hepatitis D, tis E, herpes x, histoplasmosis, hookworm infection, human rus infection, human ewingii ehrlichiosis, human granulocytic anaplasmosis (HGA), human metapneumovirus infection, Human monocytic ehrlichiosis, human papillomavirus (HPV) infection, Human parainfluenza virus infection, Hymenolepiasis, Epstein-Barr Virus ious Mononucleosis (Mono), influenza (flu), isosporiasis, kawasaki disease, keratitis, la kingae infection, kuru, lassa fever, legionellosis nnaires' disease), legionellosis (pontiac fever), leishmaniasis, leprosy, leptospirosis, listeriosis, lyme disease (lyme borreliosis), lymphatic sis antiasis), Lymphocytic choriomeningitis, Malaria, Marng hemorrhagic fever (MHF), Measles, Middle East respiratory syndrome (MERS), melioidosis (Whitmore's disease), meningitis, meningococcal e, metagonimiasis, microsporidiosis, molluscum contagiosum (MC), Monkeypox, Mumps, Murine typhus (Endemic typhus), Mycoplasma pneumonia, Mycetoma biguation), Myiasis, Neonatal conjunctivitis (Ophthalmia orum), (New) Variant Creutzfeldt-Jakob disease (vCJD, nvCJD), nocardiosis, onchocerciasis (River blindness), opisthorchiasis, ccidioidomycosis (South an blastomycosis), paragonimiasis, pasteurellosis, pediculosis capitis (head lice), losis corporis (body lice), pediculosis pubis (pubic lice, crab lice), pelvic inflammatory disease (PID), pertussis (Whooping cough), plague, pneumococcal infection, pneumocystis pneumonia (PCP), pneumonia, poliomyelitis, prevotella ion, primary amoebic meningoencephalitis (PAM), progressive multifocal leukoencephalopathy, psittacosis, Q fever, rabies, relapsing fever, respiratory syncytial virus infection, rhinosporidiosis, rhinovirus infection, rickettsial infection, rickettsialpox, Rift Valley fever (RVF), Rocky Mountain spotted fever (RMSF), rotavirus infection, rubella, salmonellosis, SARS (Severe Acute Respiratory Syndrome), scabies, schistosomiasis, sepsis, shigellosis (Bacillary dysentery), shingles (Herpes zoster), smallpox (Variola), sporotrichosis, staphylococcal food poisoning, staphylococcal infection, strongyloidiasis, subacute sclerosing ephalitis, syphilis, Taeniasis, Tetanus (Lockj aw), Tinea barbae (Barber's itch), Tinea capitis (Ringworm of the Scalp), Tinea corporis (Ringworm of the Body), Tinea cruris (Jock itch), Tinea manum (Ringworm of the Hand), Tinea nigra, Tinea pedis (Athlete’s foot), Tinea unguium (Onychomycosis), Tinea versicolor (Pityriasis versicolor), Toxocariasis (Ocular Larva Migrans (OLM)), Toxocariasis (Visceral Larva Migrans (VLM)), Trachoma, Toxoplasmosis, Trichinosis, Trichomoniasis, Trichuriasis (Whipworm infection), Tuberculosis, Tularemia, Typhoid fever, Typhus fever, Ureaplasma urealyticum infection, Valley fever, elan equine encephalitis, Venezuelan hemorrhagic fever, Vibrio vulnificus ion, Vibrio parahaemolyticus enteritis, viral pneumonia, West Nile Fever, white piedra (Tinea blanca), Yersinia pseudotuberculosis ion, yersiniosis, yellow fever, Zika fever, or zygomycosis.
Exemplary immune disorders or conditions that can be treated or prevented by the polypeptides described herein e, but are not limited to, Addison’s disease, agammaglobulinemia, alopecia areata, amyloidosis, ankylosing litis, anti-GBM/anti-TBM nephritis, antiphospholipid syndrome (APS), autoimmune hepatitis, autoimmune inner ear disease (AIED), axonal & neuronal neuropathy (AMAN), Behcet’s disease, Bullous pemphigoid, Castleman disease (CD), Celiac disease, Chagas disease, chronic inflammatory demyelinating polyneuropathy (CIDP), chronic recurrent multifocal osteomyelitis (CRMO), Churg-Strauss, icial pemphigoidfbenign mucosal pemphigoid, Cogan’s syndrome, Cold agglutinin disease, Congenital heart block, Coxsackie myocarditis, CREST me, Crohn’ s disease, itis herpetiformis, dermatomyositis, Devic’s disease (neuromyelitis ), Discoid lupus, Dressler’s syndrome, endometriosis, eosinophilic esophagitis (EoE), eosinophilic tis, erythema nodosum, essential mixed cryoglobulinemia, Evans syndrome, fibromyalgia, fibrosing alveolitis, giant cell arteritis ral arteritis), giant cell myocarditis, Glomerulonephritis, Goodpasture’s syndrome, Granulomatosis with Polyangiitis, Graves’ disease, in-Barre syndrome, oto’s thyroiditis, hemolytic anemia, Henoch-Schonlein purpura (HSP), herpes gestationis or pemphigoid gestationis (PG), hypogammalglobulinemia, IgA nephropathy, IgG4-related sclerosing disease, inclusion body myositis (IBM), interstitial cystitis (IC), le arthritis, juvenile diabetes (Type 1 diabetes), juvenile myositis (JM), Kawasaki disease, t-Eaton syndrome, leukocytoclastic vasculitis, Lichen planus, Lichen sclerosus, Ligneous conjunctivitis, linear IgA disease (LAD), lupus, Lyme disease chronic, Meniere’s e, microscopic polyangiitis (MPA), mixed connective tissue e (MCTD), Mooren’s ulcer, Mucha-Habermann disease, multiple sclerosis (MS), enia gravis, is, Narcolepsy, Neuromyelitis optica, neutropenia, ocular cicatricial pemphigoid, optic neuritis, palindromic tism (PR), PANDAS (Pediatric mune Neuropsychiatric ers Associated with Streptococcus), paraneoplastic cerebellar degeneration (PCD), smal nocturnal hemoglobinuria (PNH), Parry g syndrome, Pars planitis (peripheral uveitis), Parsonnage- Turner syndrome, Pemphigus, peripheral neuropathy, Perivenous encephalomyelitis, ious anemia (PA), POEMS syndrome (polyneuropathy, organomegaly, endocrinopathy, monoclonal gammopathy, skin changes), polyarteritis nodosa, polymyalgia rheumatica, polymyositis, postmyocardial infarction syndrome, postpericardiotomy syndrome, primary biliary cirrhosis, primary sing cholangitis, progesterone dermatitis, psoriasis, psoriatic arthritis, pure red cell aplasia (PRCA), pyoderma gangrenosum, Raynaud’s phenomenon, Reactive Arthritis, Reflex sympathetic dystrophy, Reiter’s syndrome, relapsing polychondritis, restless legs syndrome (RLS), retroperitoneal fibrosis, rheumatic fever, rheumatoid tis (RA), sarcoidosis, Schmidt syndrome, tis, scleroderma, Sj ogren’s syndrome, sperm & testicular autoimmunity, Stiff person syndrome (SPS), subacute bacterial endocarditis (SBE), Susac’s syndrome, sympathetic ophthalmia (SO), Takayasu’s arteritis, temporal arteritis/Giant cell arteritis, thrombocytopenic purpura (TTP), Tolosa-Hunt syndrome (THS), transverse myelitis, type 1 diabetes, ulcerative colitis (UC), undifferentiated connective tissue disease (UCTD), uveitis, vasculitis, vitiligo, or Wegener’s granulomatosis (Granulomatosis with Polyangiitis (GPA)).
The ptides (e.g., dy molecules or TCR molecules) described herein are typically stered at a frequency that keeps a therapeutically effective level of polypeptides in the patient’s system until the patient recovers. For example, the polypeptides may be administered at a frequency that achieves a serum tration sufficient for at least about 1, 2, 5, 10, 20, 30, or 40 polypeptides to bind each target le or cell. In an embodiment, the polypeptides are administered every 1, 2, 3, 4, 5,6, or 7 days, every 1,2,3, 4, 5, or 6 weeks, or every 1, 2, 3, 4,5, or 6 months.
Methods of administering various polypeptides (e.g., dy molecules or TCR molecules) are known in the art and are described below. Suitable dosages of the ptides used will depend on the age and weight of the subject and the particular drug used.
The polypeptides can be used by themselves or conjugated to a second agent, e.g., an bacterial agent, toxin, or protein, e.g., a second polypeptide. This method includes: administering the polypeptide, alone or conjugated to a second agent, to a subject requiring such treatment. The polypeptides can be used to deliver a variety of therapeutic agents, e.g., a toxin, or es thereof.
Combination Therapies The polypeptides (e.g., antibody molecules or TCR molecules) can be used in combination with other therapies. For example, the combination therapy can e a polypeptide co-formulated with, and/or co-administered with, one or more additional therapeutic agents, e. 3., one or more additional therapeutic agents described herein. In other embodiments, the polypeptides are administered in combination with other therapeutic treatment modalities, e.g., other therapeutic treatment modalities described . Such combination therapies may advantageously utilize lower dosages of the administered therapeutic agents, thus avoiding possible toxicities or complications associated with the various monotherapies.
Administered “in combination”, as used herein, means that two (or more) different treatments are delivered to the subject before, or during the course of the subject's affliction with a disorder. In an embodiment, two or more ents are delivered lactically, e.g., before the subject has the disorder or is diagnosed with the er. In another ment, the two or more treatments are delivered after the subject has developed or diagnosed with the disorder. In some ments, the delivery of one treatment is still occurring when the delivery of the second begins, so that there is overlap. This is mes referred to herein as "simultaneous" or "concurrent ry." In other embodiments, the delivery of one treatment ends before the delivery of the other treatment begins. In some embodiments of either case, the treatment is more effective because of combined administration.
For example, the second treatment is more effective, e.g., an equivalent effect is seen with less of the second treatment, or the second treatment reduces symptoms to a greater extent, than would be seen if the second treatment were administered in the absence of the first treatment, or the analogous situation is seen with the first treatment. In some embodiments, delivery is such that the reduction in a symptom, or other parameter related to the disorder is greater than what would be observed with one treatment delivered in the absence of the other. The effect of the two treatments can be partially ve, wholly additive, or greater than additive. The delivery can be such that an effect of the first treatment delivered is still detectable when the second is delivered.
In an embodiment, the polypeptide is administered in combination with a second therapy (e. g., an additional agent) to treat or prevent a er described herein. In an embodiment, the additional agent is a second ptide (e.g., antibody molecule), e.g., a polypeptide (e.g., an antibody molecule) different from a first ptide (e.g., antibody molecule). Exemplary polypeptides (e.g., antibody molecules) that can be used in combination include, but are not limited to, any combination of the ptides (e.g., antibody molecules) described herein. In another embodiment, the additional agent is other than a polypeptide (e.g., antibody molecule). For example, the additional agent can be a small molecule or a nucleic acid molecule. In yet another embodiment, the second therapy is chosen from a surgery, a radiation therapy, a cell therapy (e.g., a stem cell therapy), or an organ or tissue transplantation.
In an embodiment, the second therapy comprises a therapy chosen from one or more of: an androgen replacement therapy, an antihormone y, an antiserum therapy, an autologous immune enhancement therapy, a biotherapy, a blood irradiation therapy, a brachytherapy, a cardiac resynchronization therapy, a cell therapy, a cell transfer therapy, a chelation y, a herapy, a chrysotherapy, a cobalt therapy, a cold compression therapy, a cryotherapy, an electroconvulsive therapy, an electromagnetic y, an electron therapy, an electrotherapy, an enzyme replacement therapy, an epigenetic therapy, an estrogen replacement therapy, an extracorporeal shockwave therapy, a fast neutron therapy, a e therapy, a gene therapy, a heat therapy, a helminthic therapy, a hormone therapy, a hormone replacement therapy, a host modulatory therapy, a hyperbaric oxygen therapy, a hyperthermia y, an immunosuppressive therapy, an immunotherapy, an intraoperative electron ion therapy, an intraoperative radiation therapy, an inversion therapy, a laser therapy, a light y, a lithium therapy, a low level laser therapy, a magnet therapy, a magnetic resonance therapy, a medical gas y, a medical nutrition therapy, a molecular chaperone therapy, a molecular y, a monoclonal antibody therapy, a negative air ionization therapy, a n capture therapy, a neutron therapy, an oral rehydration therapy, an osmotherapy, an oxygen therapy, an ozone therapy, a palliative therapy, a particle therapy, a phage therapy, a phonemic ogical hypochromium therapy, a photodynamic therapy, a herapy, a photothermal therapy, a physical therapy, a prolotherapy, a protein therapy, a proton therapy, a pulsed electromagnetic field therapy, a PUVA y, a radiation therapy, a rehydration therapy, a respiratory therapy, salvage therapy, a serotherapy, a stem cell therapy, a stereotactic radiation therapy, a targeted therapy, a therapy, a TK cell therapy, a tolerogenic therapy, a transdermal continuous oxygen therapy, an ultraviolet light therapy, or a virotherapy.
Exemplary therapies that can be used in combination with a polypeptide or composition described herein to treat or prevent other disorders are also described in the section of “Methods of Treating or Preventing Disorders” herein.
Methods of Diagnosis In some aspects, the present disclosure provides a diagnostic method for detecting the presence of a target molecule (e.g., a n) or cell in vitro (e.g., in a biological sample, such as a biopsy or body fluid (e.g., blood, urine, or cerebrospinal fluid) sample) or in viva (e.g., in vivo imaging in a subject). The method includes: (i) contacting the sample with a polypeptide described herein (e.g., an antibody molecule described herein), or administering to the subject, the ptide (6.3., dy molecule); (optionally) (ii) contacting a reference sample, e.g., a control sample (e.g., a control biological sample, such as a biopsy or body fluid (e.g., blood, urine, or cerebrospinal fluid) ) or a control subject with a polypeptide described herein (e.g., an antibody molecule described herein); and (iii) detecting formation of a complex between the polypeptide (e. g., antibody molecule) and the target molecule or cell in the sample or subject, or the control sample or subject, wherein a change, e.g., a statistically significant change, in the formation of the complex in the sample or subject relative to the control sample or subject is indicative of the ce of the target molecule or cell in the sample. The polypeptide (e.g., antibody molecule) can be ly or indirectly labeled with a able substance to facilitate detection of the bound or unbound polypeptide (e.g., antibody molecule). le detectable substances include various enzymes, prosthetic groups, fluorescent materials, luminescent materials and radioactive materials, as described herein.
The term “sample,” as it refers to samples used for detecting bacteria includes, but is not limited to, cells, cell lysates, proteins or membrane ts of cells, body fluids such as blood, urine, or CSF, or tissue samples such as biopsies.
Complex ion n the polypeptide (e.g., antibody molecule), and the target molecule or cell, can be detected by measuring or visualizing either the polypeptide (e.g., antibody molecule) bound to the target molecule or cell, or unbound polypeptide (e.g., antibody molecule).
Any suitable detection assays can be used, and conventional detection assays include an - linked imrnunosorbent assay (ELISA), a radioimmunoassay (RIA), a FACS assay, a BIACORE assay, or tissue immunohistochemistry. Alternative to labeling the polypeptide, the presence of the target molecule or cell can be d in a sample by a ition immunoassay utilizing standards labeled with a able substance and an unlabeled polypeptide. In this assay, the biological sample, the labeled standards and the polypeptide are ed and the amount of labeled standard bound to the unlabeled binding molecule is determined. The amount of the target molecule or cell in the sample is inversely proportional to the amount of labeled standard bound to the polypeptide (e.g., antibody molecule).
The polypeptides (e.g., antibody molecules) described herein can be used to diagnose disorders that can be treated or prevented by the polypeptides described herein. The detection or diagnostic methods described herein can be used in combination with other methods described herein to treat or prevent disorders bed herein.
Additional embodiments are described in the numbered paragraphs below. 1. A method of making a nucleic acid sequence sing a sequence that encodes a heavy chain element (HC element) of an antibody heavy chain variable region (HCVR) and a light chain element (LC element) of an antibody light chain variable region (LCVR), and n the HCVR and LCVR are matched, the method comprising: a) acquiring an isolated production reaction site, e.g., a production micro-chamber, comprising: i) a heavy chain (HC) strand, wherein the HC strand is a strand of a heavy chain -stranded cDNA (HC ds cDNA) comprising a segment that encodes an HC element of the HCVR from a cell, e.g., a heavy chain variable region sequence (HCVRS); and ii) a light chain (LC) strand, wherein the LC strand is a strand of a light chain double- stranded cDNA (LC ds cDNA) comprising a segment that encodes an LC element of the LCVR from the cell, e. 3., a light chain variable region sequence (LCVRS), and b) covalent linking, e.g., ligation, of an HC strand to an LC strand, wherein the isolated production reaction site, e.g., a production micro-chamber, does not include a nucleic acid encoding an LCVR or an HCVR from a cell other than the cell, thereby making a c acid sequence comprising a sequence that encodes an HC element of an HCVR and a LC element of an LCVR, wherein the HCVR and LCVR are matched. 2. The method of paragraph 1, wherein the HC element comprises, or consists of, an HCVRS, or a functional fragment f (e.g., an antigen binding fragment thereof). 3. The method of aph 1 or 2, n the LC element comprises, or consists of, an LCVRS, or a functional fragment thereof (e.g., an antigen g fragment thereof). 4. The method of any of paragraphs 1-3, wherein the nucleic acid ce is configured such that, when expressed, the HCVRS and the LCVRS form a functional antigen binding molecule, e.g., an scFv.
. The method of paragraph 4, wherein the n binding molecule, e.g., an scFv, is functional in vitro, ex vivo, or in viva, e.g., as ined by a method or assay bed herein. 6. The method of any of aphs 1-5, wherein acquiring an isolated production reaction site, e.g., a production micro-chamber, comprises: a) acquiring a capture substrate bound to: (i) a first double-stranded cDNA (ds cDNA) comprising a strand that is complementary to a first mRNA that encodes an HCVR from a cell; and (ii) a second ds cDNA sing a strand complementary to a second mRNA encoding an LCVR from the cell (the cDNA loaded capture substrate), and b) ining the isolated production reaction site, e.g., the production micro-chamber, under conditions that allow amplification of the first and second ds cDNAs, to produce: a plurality of HC ds cDNAs comprising a segment that s an HC element of the HCVR from the cell, e.g., an HCVRS; and a plurality of LC ds cDNAs comprising a t that encodes an LC element of the LCVR from the cell, e.g., an LCVRS. 7. The method of paragraph 6, wherein the HC ds cDNA is identical, or substantially identical, to the first ds cDNA. 8. The method of paragraph 6 or 7, wherein the LC ds cDNA is identical, or substantially cal, to the second ds cDNA. 9. The method of any of paragraphs 6-8, wherein the capture substrate comprises a bead, e. g., a ic bead. 10. The method of any of paragraphs 6-9, wherein the capture substrate comprises a moiety (e.g., an oligonucleotide) which binds to cDNA, e.g., (i) a moiety which binds to the HC strand; (ii) a moiety which binds to the LC strand; or (iii) both (i) and (ii). 11. The method of any of paragraphs 6-10, wherein the first mRNA and the second mRNA are disposed on an mRNA loaded capture substrate. 12. The method of any of paragraphs 6-11, wherein the isolated production reaction site, e.g., the production micro-chamber, comprises: a reagent mixture suitable for producing, from the first and second mRNAs (e. g., after the first and second mRNAs are released from the mRNA loaded capture substrate into a solution), a first ds cDNA comprising a segment that s an HC t of the HCVR of the cell, e. g., an HCVRS, and a second ds cDNA comprising a segment that s an LC element of the LCVR of the cell, e.g., an LCVRS. 13. The method of any of paragraphs 6-12, n the isolated production reaction site, e.g., production micro-chamber, comprises primers that mediate the production of the first ds cDNA. 14. The method of any of paragraphs 6-13, wherein the isolated production reaction site, e.g., production micro-chamber, comprises primers that mediate the production of the second ds cDNA.
. The method of any of paragraphs 6-14, wherein a cDNA strand that is complementary to a first mRNA that encodes an HCVR from a cell is made by reverse transcription of the first mRNA. 16. The method of any of paragraphs 6-15, wherein a cDNA strand that is complementary to a second mRNA that encodes an LCVR from a cell is made by reverse ription of the second mRNA. 17. The method of paragraph 15 or 16, wherein the reverse transcription takes place in the isolated production reaction site, e.g., a production-micro chamber. 18. The method of paragraph 15 or 16, wherein the reverse transcription takes place in an isolated cell reaction site, e.g., a cell isolation micro-chamber. 19. The method of paragraph 15 or 16, wherein the e transcription takes place outside the isolated production reaction site, e.g., a production micro-chamber, or outside an isolated cell reaction site, e. g., a cell ion micro-chamber.
. The method of paragraph 15 or 16, wherein the reverse transcription takes place outside the isolated production reaction site, e. g., a production-micro r, and outside an isolated cell reaction site, e.g., a cell isolation micro-chamber. 21. The method of paragraph 15 or 16, wherein the reverse transcription takes place outside an isolated reaction site, e.g., outside a chamber. 22. The method of any of aphs 6-21, wherein the amplification comprises 20 or fewer , e.g., 15 or fewer, 14 or fewer, 13 or fewer, 12 or fewer, 11 or fewer, 10 or fewer, 9 or fewer, 8 or fewer, 7 or fewer, 6 or fewer, or 5 or fewer cycles. 23. The method of any of paragraphs 6-22, wherein the e transcription and/or ication uses one or more primers, e. g., comprising a sequence specific for an HCVRS and/or an LCVRS. 24. The method of any of paragraphs 6-23, wherein the amplification comprises using two or more primers that mediate the production of the HC ds cDNA, wherein at least one primer comprises a nucleotide modification, and wherein at least one primer does not comprise a nucleotide modification.
. The method of any of aphs 6-24, wherein the amplification comprises using two or more primers that mediate the tion of the LC ds cDNA, wherein at least one primer ses a nucleotide modification, and wherein at least one primer does not comprise a nucleotide modification. 26. The method of paragraph 25, wherein at least one primer comprises a nucleotide modification, e.g., which reduces, e.g., inhibits, DNA synthesis, e.g., by a DNA polymerase. 27. The method of paragraph 25 or 26, wherein at least one primer does not comprise a nucleotide modification, e.g., which reduces, e.g., inhibits, DNA synthesis, e.g., by a DNA polymerase. 28. The method of paragraph 26 or 27, wherein the nucleotide cation inhibits a DNA polymerase from extending the DNA. 29. The method of any of paragraphs 26-28, wherein the nucleotide modification is an insertion of a spacer to the , e.g., between two adjacent nucleotides in the primer.
. The method of paragraph 29, wherein the spacer is a flexible spacer, a carbon spacer (e.g., -(CH2)n-, wherein n=3, 4, 5, or more), two or more (e.g., three, four, five, or more) abasic nucleotides or a PEG spacer. 31. The method of any of paragraphs 26-28, wherein the nucleotide modification is 2’-O- methyl, 2’-OH, 2’-NH2, or uracil, e.g., to a ribose. 32. The method of any of paragraphs 26-31, wherein the nucleotide modification is located internally or at the 3’ end of the primer. 33. The method of any of paragraphs 23-32, wherein at least one primer comprises (i) a first ; (ii) a second member; and optionally (iii) a nucleotide modification described herein, e.g., located between (i) and (ii). 34. The method of paragraph 33, wherein the first member is e of annealing with the second member in the same primer or a different primer, e.g., forming a hairpin structure (via intramolecular hybridization) or a -stranded structure (via intermolecular ization) comprising a duplex region of 4, 5, 6, 7, 8, 9, 10, ll, 12, 13, 14, 15, 16, 17, 18, 19, 20, more basepairs.
. The method of paragraph 33 or 34, wherein the first member comprises a sequence that is mentary to the sequence of an oligonucleotide attached to the capture substrate. 36. The method of any of paragraphs 33-35, wherein the second member comprises (e.g., from 5’ to 3’) one, two, or all of: (i) a sequence that is complementary to at least a portion of the first member; (ii) a universal priming sequence (e.g., for PCR amplification or next-generation sequencing); and (iii) a sequence complementary to a target sequence, e.g., an HCVRS and/or a LCVRS, ally, wherein the second member ses a sequence for homologous recombination (e.g., in a yeast or mammalian cell). 37. The method of any of paragraphs 23-36, n at least one primer comprises a sequence encoding at least a portion of a linker sequence, or a complementary sequence thereof. 38. The method of paragraph 37, wherein the primer that comprises a sequence encoding at least a portion of a linker sequence, or a complementary sequence thereof, is orylated, e.g., 5’ phosphorylated. 39. The method of paragraph 37 or 38, wherein the linker sequence comprises, or consists of, (Gly-Gly-Gly-Gly-Ser)n, where n=1, 2, 3, 4, 5, or more. 40. The method of any of paragraphs 1-39, n the HC ds cDNA comprises a 5’ overhang, e.g., a 5’ overhang that is capable of hybridizing to an oligonucleotide attached to a capture substrate. 41. The method of any of paragraphs 1-40, wherein the HC ds cDNA comprises a blunt end, e.g., a blunt end comprising a 5’ phosphate. 42. The method of any of paragraphs 1-41, n the LC ds cDNA comprises a 5’ overhang, e.g., a 5’ overhang that is capable of hybridizing to an ucleotide attached to a capture substrate. 43. The method of any of paragraphs 1-42, wherein the LC ds cDNA comprises a blunt end, e.g., a blunt end comprising a 5’ phosphate. 44. The method of any of paragraphs 1-43, wherein the HC ds cDNA and the LC ds cDNA comprise sticky ends, e.g., both have 5’ overhangs. 45. The method of any of paragraphs 1-44, wherein the HC strand and the LC strand are covalently linked, e.g., d, to produce a single stranded nucleic acid sequence, wherein the HC and LC strands are both sense strands or both antisense strands. 46. The method of any of paragraphs 1-44, wherein a denatured HC strand of the HC ds cDNA to a denatured LC strand of the LC ds cDNA are covalently linked, e.g., ligated, wherein the HC and LC strands are both sense strands or both antisense strands. 47. The method of any of paragraphs 1-44, wherein the HC strand is present in the HC ds cDNA and the LC strand is present in the LC ds cDNA, and wherein the HC ds cDNA and the LC ds cDNA are covalently linked, e.g., ligated, e.g., to produce a double stranded nucleic acid sequence. 48. The method of any of paragraphs 1-47, wherein the covalent g, e.g., ligation, occurs in the isolated production reaction site. 49. The method of paragraph 48, wherein the isolated production reaction site, e.g., a production micro-chamber, ses a reagent that is capable of covalently linking, e.g., ligating, the HC and LC strands or the HC and LC ds cDNAs. 50. The method of aph 48 or 49, wherein the isolated tion reaction site, e.g., a production micro-chamber comprises an enzyme that covalently couples the HC and LC s or the HC and LC ds cDNAs. 51. The method of any of aphs 1-47, wherein the covalent linking, e.g., ligation, occurs in a site different from the isolated production reaction site, e.g., occurs in an isolated linkage reaction site, e.g., a linkage micro-chamber. 52. The method of paragraph 51, wherein the HC strand and the LC strand are transferred from the isolated production site to the isolated linkage reaction site, e.g., a e micro-chamber, and the covalent linking occurs in the isolated linkage reaction site, e.g., a e micro-chamber. 53. The method of paragraph 51 or 52, n the isolated linkage reaction site, e.g., a linkage micro-chamber, comprises a reagent that is capable of covalently linking, e.g., ligating, the HC and LC strands or the HC and LC ds cDNAs. 54. The method of any of paragraphs 51-53, wherein the isolated linkage reaction site, e. g., a linkage micro-chamber, ses an enzyme that covalently couples the HC and LC strands or the HC and LC ds cDNAs. 55. The method of aph 50 or 54, wherein the enzyme is a ligase, e.g., a thermal stable ligase. 56. The method of any of paragraphs 51-55, wherein the covalent linking, e.g., on, comprises: (a) heating the isolated linkage on site, e.g., the linkage micro-chamber, under conditions (e.g., at 95°C) that allow denaturation of the HC strand and the LC strand; (b) cooling the isolated linkage reaction site, e.g., the linkage micro-chamber, under conditions (e.g., at 50-65°C) that allow hybridization of the splint oligonucleotide to the HC strand and the LC strand; (c) maintaining the isolated linkage reaction site, e.g., the linkage micro-chamber, under conditions (e.g., at 45-65°C) that allow ligation of the HC strand and the LC strand (e.g., formation of phosphodiester bond between the HC strand and the LC strand); and (d) repeating steps (a), (b), and (c) sequentially for 2, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, or more cycles. 57. The method of any of paragraphs 1-56, wherein the HC strand and the LC strand are covalently linked, e.g., ligated, in the ce of a splint oligonucleotide. 58. The method of paragraph 57, wherein the splint oligonucleotide is hybridized to a sequence comprising the junction of the HC strand and the DC , or a sequence complementary thereof, and forms a duplexed region at the site of ligation. 59. The method of paragraph 57 or 58, wherein the splint oligonucleotide comprises a modification (e.g., an NH2 group) that inhibits DNA synthesis, e.g., by a DNA polymerase. 60. The method of paragraph 59, wherein the modification is at the 3’ end of the splint ucleotide. 61. The method of any of paragraphs 1-60, n a strand complimentary to the covalently linked, e.g., ligated, HC and LC strands is produced by ication. 62. The method of any of aphs 1-61, further comprising, prior to acquiring the isolated production reaction site, e.g., a production micro-chamber, acquiring an mRNA loaded capture substrate. 63. The method of paragraph 62, n acquiring the mRNA loaded capture substrate comprising: a) acquiring an ed cell reaction site, e.g., a cell isolation micro-chamber, comprising: i) a cell; and ii) a capture substrate capable of binding a first mRNA encoding an HCVR from the cell and a second mRNA ng an LCVR from the cell; and b) maintaining the isolated cell reaction site, e.g., the cell isolation chamber, under conditions that allow lysis of the cell and binding of the capture substrate with the first mRNA and the second mRNA to form the mRNA loaded capture substrate, wherein the isolated cell reaction site, e.g., cell isolation micro-chamber, does not include a nucleic acid ng an HCVR or an LCVR from a cell other than the cell. 64. The method of paragraph 63, wherein the isolated cell on site, e. g., cell isolation micro-chamber, comprises a lysing reagent, e.g., a detergent. 65. The method of paragraph 63 or 64, wherein the cell is lysed by heat or an enzyme. 66. The method of any of paragraphs 63-65, wherein the capture substrate comprises a moiety (e.g., an oligonucleotide) which binds mRNA, e.g., an oligo(dT). 67. The method of any of paragraphs 62-66, further comprising releasing the mRNA loaded capture substrate from the isolated cell reaction site, e.g., the cell isolation micro-chamber. 68. The method of paragraph 67, n the releasing step is performed in the presence of a poly(dA) or poly(dT) oligonucleotide, e.g., to reduce cross-binding of non-captured mRNA. 69. The method of paragraph 62-68, wherein the mRNA loaded capture substrate is transferred from the isolated cell reaction site, e.g., the cell ion micro-chamber, to the isolated production reaction site, e.g., the production micro-chamber. 70. The method of any of paragraphs 1-69, comprising releasing the c acid sequence from the production micro-chamber. 71. The method of any of paragraphs 1-70, further comprising amplifying the nucleic acid sequence. 72. The method of any of paragraphs 1-71, comprising sequencing all or a portion of the c acid sequence. 73. The method of any of paragraphs 1-72, comprising inserting all or a portion of c acid sequence into a vector. 74. The method of aph 73, wherein the vector supplies an additional HC element or LC element not included in the nucleic acid sequence. 75. The method of paragraph 73 or 74, n the vector supplies an HC CDRl, an HC CDRZ, or both. 76. The method of any of paragraphs 73-75, comprising expressing the . 77. The method of any of paragraphs 1-76, comprising sing the c acid sequence to produce a polypeptide comprising a segment that encodes an HC element of the HCVR, e.g., an HCVRS, and a segment that encodes an LC element of the LCVR, e.g., an LCVRS. 78. The method of paragraph 77, wherein the LC element is N-terminal to the HC element in the ptide. 79. The method of paragraph 77, wherein the HC element is C-terminal to the LC element in the polypeptide. 80. The method of any of paragraphs 77-79, further comprising contacting the polypeptide with an antigen. 81. The method of any of aphs 77-80, further comprising determining if the polypeptide binds the antigen. 82. The method of any of paragraphs 1-81, wherein the cell is an immune cell, e.g., a B cell or T cell, e. g., a human B cell or T cell. 83. The method of any of paragraphs 1-82, wherein the cell is a mammalian cell or an avian cell. 84. A method of making a nucleic acid sequence comprising a sequence that s a heavy chain element (HC element) of an antibody heavy chain variable region (HCVR) and a light chain element (LC element) of an antibody light chain variable region (LCVR), and wherein the HCVR and LCVR are matched, comprising: a) acquiring an isolated cell reaction site, e.g., a cell isolation micro-chamber, comprising: i) a cell; and ii) a capture substrate capable of binding a first mRNA encoding an HCVR from the cell and a second mRNA ng an LCVR from the cell; b) maintaining isolated cell reaction site, e.g., the cell isolation micro-chamber, under conditions that allow lysis of the cell and binding of the e substrate with the first mRNA and the second mRNA to form the mRNA loaded capture ate, wherein the isolated cell reaction site, e.g., cell isolation micro-chamber, does not include a nucleic acid encoding an HCVR or an LCVR from a cell other than the cell; c) contacting the mRNA loaded capture substrate with a reaction mixture, e.g., a reaction mixture comprising reverse transcriptase, that uses the loaded mRNA as a template to make cDNA (this can occur, e.g., in the isolated cell on site, in the isolated production reaction site, or in neither, e.g., not in an ed reaction site); d) acquiring an isolated production reaction site, e.g., a production micro-chamber, comprising: i) a heavy chain (HC) strand from step b), wherein the HC strand is a strand of a heavy chain double-stranded cDNA (HC ds cDNA) comprising a segment that encodes an HC element of the HCVR from the cell, e.g., a heavy chain variable region sequence (HCVRS); ii) a light chain (LC) strand from step b), wherein the LC strand is a strand of a light chain -stranded cDNA (LC ds cDNA) comprising a segment that encodes an LC element of the LCVR from the cell, e.g., a light chain variable region sequence (LCVRS); and e) covalent linking, e.g., on, of an HC strand to an LC strand, wherein the isolated production on site, e.g., a production micro-chamber, does not include a nucleic acid encoding an LCVR or an HCVR from a cell other than the cell. 85. A method of making a nucleic acid sequence comprising a sequence that encodes a heavy chain element (HC t) of an antibody heavy chain le region (HCVR) and a light chain element (LC element) of an antibody light chain variable region , and n the HCVR and LCVR are matched, comprising: a) acquiring an isolated cell reaction site, e.g., a cell isolation micro-chamber, comprising: i) a cell; and ii) a capture substrate capable of binding a first mRNA encoding an HCVR from the cell and a second mRNA encoding an LCVR from the cell; b) maintaining the isolated cell reaction site, e.g., the cell isolation chamber, under conditions that allow lysis of the cell and binding of the capture substrate with the first mRNA and the second mRNA to form the mRNA loaded capture substrate, wherein the isolated cell reaction site, e.g., cell isolation micro-chamber, does not include a nucleic acid encoding an HCVR or an LCVR from a cell other than the cell; c) acquiring an isolated production reaction site, e.g., a tion micro-chamber, comprises: contacting the mRNA loaded capture substrate with a reaction mixture, e.g., a reaction mixture comprising reverse transcriptase, that uses the loaded mRNA as a template, to produce: a first double-stranded cDNA (ds cDNA) comprising a strand that is mentary to a first mRNA that encodes an HCVR from a cell; and a second ds cDNA comprising a strand complementary to a second mRNA encoding an LCVR from the cell (the cDNA loaded e substrate); wherein the isolated production reaction site, e.g., a production micro-chamber, does not include a nucleic acid encoding an LCVR or an HCVR from a cell other than the cell. d) ining the isolated production reaction site, e.g., the production micro-chamber, under conditions that allow amplification of the first and second ds cDNAs, to produce: a plurality of HC ds cDNAs comprising a segment that encodes an HC element of the HCVR from the cell, e.g., an HCVRS; and a plurality of LC ds cDNAs comprising a t that encodes an LC element of the LCVR from the cell, e. g., an LCVRS; e) acquiring an isolated linkage reaction site, e.g., a linkage micro-chamber, comprising: covalent linking, e.g., ligation, of a strand of the HC ds cDNA (HC strand) to a strand of the LC ds cDNA (LC strand), wherein the HC and LC strands are both sense strands or nse strands; and f) amplifying the covalently linked, e.g., ligated, HC and LC strands. 86. A method of making a library comprising a ity of unique members, the method comprising: making the plurality of members, wherein each of the members comprises a sequence that encodes a heavy chain t (HC element) of a heavy chain variable region (HCVR) and a light chain element (LC element) of a light chain variable region , and wherein the HCVR and LCVR are matched, made by a method of any of paragraphs 1-85, wherein each unique nucleic acid ce of the plurality comprises an HC element and an LC element from a different unique cell, thereby making a library comprising a plurality of unique members. 87. The method of paragraph 86, wherein the ity of unique members ses at least 104, 105, 106, 107, 108, or 109 unique members. 88. The method of paragraph 86 or 87, wherein the plurality of unique members comprises 104 to 109, 104 to 108, 104 to 107, 104 to 106, 104 to 105, 108 to 109, 107 to 109, 106 to 109, 105 to 109, 105 to 108, 106 to 107, 104 to 105, 105 to 106, 106 to 107, 107 to 108, or 108 to 109 unique s. 89. The method of any of paragraphs 86-88, wherein at least 80%, 85 %, 90%, 95%, 96%, 97%, 98%, 99%, or 100%, of the s in the library are unique members (which encode matched HC element and LC element sequences). 90. The method of any of paragraphs 86-89, wherein less than 20%, 15%, 10%, 5%, 4%, 3%, 2%, or 1%, of the members in the library are unique members (which encode matched HC element and LC element sequences). 91. A library comprising: a plurality of unique members, wherein, i) each unique member of the plurality comprises a segment that encodes an HC element, e.g., an HCVRS, and a segment that encodes an LC element, e.g., an LCVRS, wherein the HC element and the LC element in each unique member is matched; ii) each unique member of the plurality comprises a segment that encodes an HC element, e.g., an HCVRS, and a segment that encodes an LC element, e.g., an LCVRS, from a different unique cell; and iii) the library ses one or more (e.g., two, three, four, or all) of the following properties: a) the library is made by a method of any of paragraphs 1-85; b) the plurality of unique members comprises at least 104, 105, 106, 107, 108, or 109 unique nucleic acid sequences; c) the plurality of unique members comprises 104 to 109, 104 to 108, 104 to 107, 104 to 106, 104 to 105, 108 to 109, 107 to 109, 106 to 109, 105 to 109, 105 to 108, 106 to 107, 104 to 105, 105 to 106, 106 to 107, 107 to 108, or 108 to 109 unique members; (1) at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%, of the members in the y are unique members (which encode matched HC element and LC element ces); or e) less than 20%, 15%, 10%, 5%, 4%, 3%, 2%, or 1%, of the members in the library are unique members (which encode matched HC element and LC element sequences). 92. The library of aph 91, wherein each unique member of the plurality is configured such that, when expressed, the HC element, e.g., the HCVRS, and the LC element, e. g., the LCVRS, form a functional antigen binding molecule, e.g., an scFV. 93. The y of any of paragraphs 91-92, wherein the library is a display library. 94. The library of any of paragraphs 91-93, wherein each of the members of the plurality r s a polypeptide that s in display of the member on the e of a display entity. 95. The library of any of paragraphs 91-94, wherein the library is a phage display library. 96. The library of any of paragraphs 91-94, wherein the library is a yeast display library. 97. The y of any of paragraphs 91-94, wherein the y is a mammalian y library. 98. A method of making a binding polypeptide, the method comprising: a) acquiring a y of any of paragraphs 91-97; and b) expressing a polypeptide d by a unique nucleic acid of the library. 99. The method of paragraph 98, further comprising contacting the polypeptide with an antigen. 100. The method of paragraph 98 or 99, further comprising retrieving the nucleic acid that encodes a polypeptide that binds the antigen. 101. A method of making a nucleic acid sequence comprising a sequence that encodes an 0: chain element (AC element) of a TCR 0t chain variable region (ACVR) and a [3 chain element (BC element) of a TCR [3 chain le region (BCVR), and wherein the ACVR and BCVR are d, the method sing: a) ing an isolated production reaction site, e.g., a production micro-chamber, comprising: i) an a chain (AC) , wherein the AC strand is a strand of an 0. chain - stranded cDNA (AC ds cDNA) comprising a segment that encodes an AC element of the ACVR from a cell, e.g., an a chain le region sequence (ACVRS); and ii) a [5 chain (BC) strand, wherein the BC strand is a strand of a B chain double- stranded cDNA (BC ds cDNA) comprising a segment that encodes a BC element of the BCVR from the cell, e.g., a [3 chain variable region ce (BCVRS), and b) covalent linking, e.g., ligation, of an AC strand to a BC strand, wherein the isolated production reaction site, e.g., a production micro-chamber, does not include a nucleic acid encoding a BCVR or an ACVR from a cell other than the cell, thereby making a nucleic acid sequence comprising a sequence that encodes an AC element of an ACVR and a BC element of a BCVR, wherein the ACVR and BCVR are matched. 102. The method of paragraph 101, wherein the AC element comprises, or consists of, an ACVRS, or a functional fragment thereof (e.g., an antigen binding fragment thereof). 103. The method of paragraph 101 or 102, wherein the BC element comprises, or consists of, a BCVRS, or a functional fragment thereof (e.g., an antigen binding fragment thereof). 104. The method of any of paragraphs 101-103, wherein the nucleic acid sequence is red such that, when expressed, the ACVRS and the BCVRS form a functional antigen binding molecule, e. g., a single chain TCR molecule. 105. The method of paragraph 104, wherein the antigen binding molecule is functional in vitro, ex vivo, or in viva, e.g., as determined by a method or assay described herein. 106. The method of any of paragraphs 101-105, wherein acquiring an ed production reaction site, e.g., a production micro-chamber, comprises: a) acquiring a capture ate bound to: (i) a first double-stranded cDNA (ds cDNA) comprising a strand that is complementary to a first mRNA that encodes an ACVR from a cell; and (ii) a second ds cDNA comprising a strand complementary to a second mRNA encoding a BCVR from the cell (the cDNA loaded capture substrate), and b) ining the isolated tion reaction site, e.g., the production micro-chamber, under conditions that allow cation of the first and second ds cDNAs, to produce: a plurality of AC ds cDNAs comprising a segment that encodes an AC element of the ACVR from the cell, e.g., an ACVRS; and a plurality of BC ds cDNAs comprising a segment that encodes a BC element of the BCVR from the cell, e.g., a BCVRS. 107. The method of paragraph 106, wherein the AC ds cDNA is identical, or substantially identical, to the first ds cDNA. 108. The method of paragraph 106 or 107, n the BC ds cDNA is identical, or substantially identical, to the second ds cDNA. 109. The method of any of paragraphs 106-108, wherein the e substrate comprises a bead, e.g., a magnetic bead. 110. The method of any of paragraphs 106-109, wherein the capture substrate comprises a moiety (e.g., an ucleotide) which binds to cDNA, e.g., (i) a moiety which binds to the AC strand; (ii) a moiety which binds to the BC strand; or (iii) both (i) and (ii). 111. The method of any of paragraphs 106-110, wherein the first mRNA and the second mRNA are disposed on an mRNA loaded capture ate. 112. The method of any of paragraphs 106-111, wherein the isolated production reaction site, e.g., the production micro-chamber, comprises: a reagent mixture suitable for producing, from the first and second mRNAs (e.g., after the first and second mRNAs are released from the mRNA loaded capture substrate into a solution), a first ds cDNA comprising a segment that encodes an AC element of the ACVR of the cell, e. g., an ACVRS, and a second ds cDNA comprising a segment that encodes a BC element of the BCVR of the cell, e.g., a BCVRS. 113. The method of any of aphs 106-112, wherein the isolated tion reaction site, e.g., production micro-chamber, comprises primers that mediate the production of the first ds cDNA. 114. The method of any of paragraphs 106-113, wherein the isolated production reaction site, e.g., production micro-chamber, comprises primers that mediate the production of the second ds cDNA.
WO 19402 115. The method of any of paragraphs 106-114, wherein a cDNA strand that is complementary to a first mRNA that encodes an ACVR from a cell is made by reverse transcription of the first mRNA. 116. The method of any of paragraphs 106-115, n a cDNA strand that is complementary to a second mRNA that encodes a BCVR from a cell is made by reverse transcription of the second mRNA. 117. The method of paragraph 115 or 116, wherein the reverse transcription takes place in the isolated production reaction site, e.g., a production-micro chamber. 118. The method of paragraph 115 or 116, wherein the reverse transcription takes place in an isolated cell reaction site, e.g., a cell isolation micro-chamber. 119. The method of paragraph 115 or 116, wherein the reverse transcription takes place outside the isolated production reaction site, e.g., a production micro-chamber, or outside an isolated cell reaction site, e.g., a cell isolation micro-chamber. 120. The method of paragraph 115 or 116, wherein the e transcription takes place outside the isolated production on site, e.g., a production-micro chamber, and outside an isolated cell reaction site, e.g., a cell ion micro-chamber. 121. The method of paragraph 115 or 116, wherein the reverse ription takes place outside an ed reaction site, e.g., e a micro-chamber. 122. The method of any of paragraphs 106-121, n the amplification comprises 20 or fewer cycles, e.g., 15 or fewer, 14 or fewer, 13 or fewer, 12 or fewer, 11 or fewer, 10 or fewer, 9 or fewer, 8 or fewer, 7 or fewer, 6 or fewer, or 5 or fewer cycles. 123. The method of any of paragraphs 106-122, wherein the reverse transcription and/or amplification uses one or more primers, e.g., comprising a sequence specific for an ACVRS and/or a BCVRS. 124. The method of any of paragraphs 106-123, n the amplification comprises using two or more primers that mediate the production of the AC ds cDNA, wherein at least one primer comprises a nucleotide modification, and wherein at least one primer does not comprise a nucleotide modification. 125. The method of any of paragraphs 106-124, wherein the amplification comprises using two or more primers that mediate the production of the BC ds cDNA, wherein at least one primer comprises a nucleotide modification, and n at least one primer does not comprise a nucleotide modification. 126. The method of aph 125, wherein at least one primer comprises a nucleotide modification, e.g., which reduces, e.g., inhibits, DNA synthesis, e.g., by a DNA polymerase. 127. The method of paragraph 125 or 126, wherein at least one primer does not comprise a nucleotide modification, e.g., which reduces, e.g., inhibits, DNA synthesis, e.g., by a DNA polymerase. 128. The method of paragraph 126 or 127, wherein the nucleotide cation inhibits a DNA polymerase from extending the DNA. 129. The method of any of paragraphs 126-128, wherein the nucleotide modification is an insertion of a spacer to the primer, e.g., between two adjacent nucleotides in the primer. 130. The method of paragraph 129, wherein the spacer is a flexible spacer, a carbon spacer (e.g., -(CH2)n-, wherein n=3, 4, 5, or more), two or more (e.g., three, four, five, or more) abasic nucleotides or a PEG . 131. The method of any of paragraphs 8, wherein the nucleotide modification is 2’-O- methyl, 2’-OH, 2’-NH2, or uracil, e.g., to a ribose. 132. The method of any of aphs 126-131, wherein the nucleotide ation is located internally or at the 3’ end of the primer. 133. The method of any of paragraphs 123-132, wherein at least one primer comprises (i) a first member; (ii) a second member; and optionally (iii) a nucleotide modification described herein, e. g., located between (i) and (ii). 134. The method of paragraph 133, wherein the first member is capable of ing with the second member in the same primer or a different primer, e.g., forming a n structure (via intramolecular hybridization) or a double-stranded structure (via intermolecular hybridization) comprising a duplex region of 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, more basepairs. 135. The method of paragraph 133 or 134, wherein the first member comprises a sequence that is complementary to the sequence of an oligonucleotide attached to the capture substrate. 136. The method of any of paragraphs 133-135, wherein the second member ses (e.g., from 5’ to 3’) one, two, or all of: (i) a sequence that is complementary to at least a n of the first member; (ii) a universal priming sequence (e.g., for PCR amplification or next-generation cing); and (iii) a sequence complementary to a target sequence, e. g., an ACVRS and/or a BCVRS, optionally, wherein the second member comprises a sequence for homologous recombination (e.g., in a yeast or mammalian cell). 137. The method of any of paragraphs 123-136, wherein at least one primer comprises a sequence encoding at least a portion of a linker sequence, or a mentary ce thereof. 138. The method of paragraph 137, wherein the primer that comprises a sequence encoding at least a n of a linker sequence, or a complementary sequence thereof, is phosphorylated, e.g., ’ phosphorylated. 139. The method of paragraph 137 or 138, wherein the linker sequence ses, or consists of, (Gly-Gly-Gly-Gly-Ser)n, where n=1, 2, 3, 4, 5, or more. 140. The method of any of paragraphs 101-139, wherein the AC ds cDNA comprises a 5’ overhang, e.g., a 5’ ng that is capable of hybridizing to an oligonucleotide attached to a capture substrate. 141. The method of any of paragraphs 101-140, wherein the AC ds cDNA comprises a blunt end, e.g., a blunt end comprising a 5’ phosphate. 142. The method of any of paragraphs 101-141, wherein the BC ds cDNA comprises a 5’ overhang, e.g., a 5’ overhang that is capable of hybridizing to an oligonucleotide attached to a capture substrate. 143. The method of any of paragraphs 2, wherein the BC ds cDNA comprises a blunt end, e.g., a blunt end comprising a 5’ phosphate. 144. The method of any of paragraphs 101-143, wherein the AC ds cDNA and the BC ds cDNA comprise sticky ends, e.g., both have 5’ overhangs. 145. The method of any of paragraphs 101-144, wherein the AC strand and the BC strand are covalently linked, e.g., ligated, to produce a single stranded nucleic acid sequence, wherein the AC and BC strands are both sense strands or both antisense strands. 146. The method of any of aphs 101-144, wherein a denatured AC strand of the AC ds cDNA to a denatured BC strand of the BC ds cDNA are ntly linked, e.g., ligated, wherein the AC and BC strands are both sense strands or both antisense strands. 147. The method of any of aphs 101-144, wherein the AC strand is present in the AC ds cDNA and the BC strand is present in the BC ds cDNA, and wherein the AC ds cDNA and the BC ds cDNA are covalently linked, e.g., ligated, e. g., to produce a double stranded nucleic acid sequence. 148. The method of any of paragraphs 101-147, wherein the covalent linking, e.g., ligation, occurs in the isolated production reaction site. 149. The method of paragraph 148, wherein the isolated production on site, e.g., a production micro-chamber, ses a reagent that is capable of covalently linking, e. g., ligating, the AC and BC s or the AC and BC ds cDNAs. 150. The method of paragraph 148 or 149, wherein the isolated production reaction site, e.g., a production chamber comprises an enzyme that covalently couples the AC and BC strands or the AC and BC ds cDNAs. 151. The method of any of paragraphs 101-147, wherein the covalent linking, e. g., ligation, occurs in a site different from the isolated production reaction site, e.g., occurs in an isolated e reaction site, e.g., a linkage chamber. 152. The method of paragraph 151, wherein the AC strand and the BC strand are transferred from the isolated production site to the isolated linkage reaction site, e.g., a e micro-chamber, and the covalent linking occurs in the isolated linkage reaction site, e.g., a linkage micro-chamber. 153. The method of paragraph 151 or 152, wherein the isolated linkage reaction site, e.g., a linkage chamber, ses a reagent that is capable of covalently linking, e.g., ligating, the AC and BC strands or the AC and BC ds cDNAs.
WO 19402 154. The method of any of paragraphs 151-153, wherein the ed linkage reaction site, e.g., a linkage micro-chamber, comprises an enzyme that covalently couples the AC and BC strands or the AC and BC ds cDNAs. 155. The method of paragraph 150 or 154, wherein the enzyme is a ligase, e.g., a l stable ligase. 156. The method of any of aphs 151-155, wherein the covalent linking, e.g., ligation, comprises: (a) heating the isolated linkage reaction site, e.g., the e micro-chamber, under conditions (e.g., at 95°C) that allow denaturation of the AC strand and the BC strand; (b) cooling the isolated linkage reaction site, e.g., the linkage micro-chamber, under conditions (e.g., at C) that allow ization of the splint oligonucleotide to the AC strand and the BC strand; (c) maintaining the isolated linkage reaction site, e.g., the linkage chamber, under conditions (e.g., at 45-65°C) that allow ligation of the AC strand and the BC strand (e.g., formation of phosphodiester bond n the AC strand and the BC strand); and (d) repeating steps (a), (b), and (c) sequentially for 2, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, or more cycles. 157. The method of any of paragraphs 101-156, wherein the AC strand and the BC strand are covalently linked, e.g., ligated, in the presence of a splint oligonucleotide. 158. The method of paragraph 157, wherein the splint oligonucleotide is hybridized to a sequence comprising the junction of the AC strand and the BC strand, or a sequence complementary thereof, and forms a duplexed region at the site of ligation. 159. The method of paragraph 157 or 158, wherein the splint oligonucleotide comprises a modification (e.g., an NH2 group) that inhibits DNA synthesis, e.g., by a DNA polymerase. 160. The method of paragraph 159, wherein the modification is at the 3’ end of the splint oligonucleotide. 161. The method of any of paragraphs 101-160, wherein a strand complimentary to the covalently linked, e.g., ligated, AC and BC s is produced by amplification. 162. The method of any of paragraphs 101-161, further comprising, prior to ing the isolated production reaction site, e.g., a production micro-chamber, acquiring an mRNA loaded capture substrate. 163. The method of paragraph 162, wherein ing the mRNA loaded capture substrate comprising: a) acquiring an isolated cell reaction site, e.g., a cell isolation micro-chamber, sing: i) a cell; and ii) a capture substrate capable of binding a first mRNA encoding an ACVR from the cell and a second mRNA encoding a BCVR from the cell; and b) maintaining the isolated cell reaction site, e.g., the cell isolation micro-chamber, under conditions that allow lysis of the cell and binding of the capture substrate with the first mRNA and the second mRNA to form the mRNA loaded capture substrate, wherein the isolated cell reaction site, e.g., cell isolation micro-chamber, does not include a nucleic acid encoding an ACVR or a BCVR from a cell other than the cell. 164. The method of paragraph 163, wherein the isolated cell reaction site, e.g., cell isolation micro-chamber, comprises a lysing reagent, e.g., a detergent. 165. The method of paragraph 163 or 164, wherein the cell is lysed by heat or an enzyme. 166. The method of any of paragraphs 163-165, wherein the capture ate comprises a moiety (e.g., an oligonucleotide) which binds mRNA, e.g., an oligo(dT). 167. The method of any of paragraphs 162-166, r comprising releasing the mRNA loaded e substrate from the isolated cell reaction site, e.g., the cell isolation micro-chamber. 168. The method of paragraph 167, wherein the releasing step is performed in the presence of a poly(dA) or poly(dT) oligonucleotide, e.g., to reduce cross-binding of non-captured mRNA. 169. The method of aph 162-168, wherein the mRNA loaded capture substrate is transferred from the isolated cell reaction site, e.g., the cell isolation micro-chamber, to the isolated production on site, e.g., the production micro-chamber. 170. The method of any of paragraphs 101-169, comprising releasing the nucleic acid sequence from the production chamber. 171. The method of any of paragraphs 101-170, further comprising ying the nucleic acid sequence. 172. The method of any of paragraphs 101-171, comprising sequencing all or a n of the c acid sequence. 173. The method of any of paragraphs , comprising inserting all or a portion of nucleic acid sequence into a . 174. The method of paragraph 173, wherein the vector supplies an onal AC element or BC element not included in the nucleic acid sequence. 175. The method of paragraph 173 or 174, wherein the vector supplies an AC CDRl, an AC CDR2, or both. 176. The method of any of paragraphs 173-175, comprising expressing the vector. 177. The method of any of aphs 101-176, comprising sing the nucleic acid sequence to produce a polypeptide comprising a segment that encodes an AC element of the ACVR, e. g., an ACVRS, and a segment that encodes a BC element of the BCVR, e. g., a BCVRS. 178. The method of paragraph 177, wherein the BC element is N—terminal to the AC element in the polypeptide. 179. The method of paragraph 177, wherein the AC element is C-terminal to the BC element in the polypeptide. 180. The method of any of paragraphs 177-179, further comprising contacting the polypeptide with an antigen. 181. The method of any of paragraphs 177-180, further comprising ining if the polypeptide binds the antigen. 182. The method of any of paragraphs 101-181, wherein the cell is an immune cell, e.g., a B cell or T cell, e.g., a human B cell or T cell. 183. The method of any of paragraphs 101-182, wherein the cell is a ian cell or an avian cell. 184. A method of making a nucleic acid sequence comprising a sequence that encodes an 0: chain element (AC element) of a TCR 0t chain variable region (ACVR) and a [3 chain element (BC element) of a TCR [3 chain le region (BCVR), and wherein the ACVR and BCVR are matched, comprising: a) acquiring an isolated cell reaction site, e.g., a cell isolation micro-chamber, comprising: i) a cell; and ii) a capture substrate capable of g a first mRNA encoding an ACVR from the cell and a second mRNA encoding a BCVR from the cell; b) maintaining isolated cell reaction site, e.g., the cell isolation micro-chamber, under conditions that allow lysis of the cell and binding of the capture substrate with the first mRNA and the second mRNA to form the mRNA loaded capture substrate, wherein the isolated cell reaction site, e. g., cell isolation micro-chamber, does not include a nucleic acid encoding an ACVR or a BCVR from a cell other than the cell; c) contacting the mRNA loaded capture ate with a reaction mixture, e.g., a reaction mixture comprising reverse transcriptase, that uses the loaded mRNA as a template to make cDNA (this can occur, e.g., in the ed cell reaction site, in the ed production reaction site, or in neither, e.g., not in an isolated reaction site); d) acquiring an isolated production reaction site, e.g., a production chamber, sing: i) an on chain (AC) strand from step b), wherein the AC strand is a strand of an 0t chain double-stranded cDNA (AC ds cDNA) sing a segment that encodes an AC element of the ACVR from the cell, e.g., a 0L chain variable region sequence (ACVRS); and ii) a [3 chain (BC) strand from step b), wherein the BC strand is a strand of a [3 chain double-stranded cDNA (BC ds cDNA) comprising a segment that encodes a BC element of the BCVR from the cell, e.g., a B chain variable region sequence (BCVRS); and e) covalent linking, e.g., ligation, of an AC strand to a BC , wherein the isolated production reaction site, e.g., a production micro-chamber, does not e a nucleic acid encoding a BCVR or an ACVR from a cell other than the cell. 185. A method of making a nucleic acid sequence comprising a sequence that s a or chain element (AC element) of an TCR or chain le region (ACVR) and a [3 chain element (BC element) of an TCR B chain variable region (BCVR), and wherein the ACVR and BCVR are d, comprising: a) acquiring an isolated cell reaction site, e.g., a cell isolation chamber, comprising: i) a cell; and ii) a capture substrate capable of binding a first mRNA encoding an ACVR from the cell and a second mRNA encoding a BCVR from the cell; b) maintaining the isolated cell reaction site, e.g., the cell ion micro-chamber, under conditions that allow lysis of the cell and binding of the capture ate with the first mRNA and the second mRNA to form the mRNA loaded capture substrate, wherein the isolated cell on site, e.g., cell isolation micro-chamber, does not include a nucleic acid encoding an ACVR or a BCVR from a cell other than the cell; c) acquiring an isolated production reaction site, e. g., a production micro-chamber, comprises: ting the mRNA loaded capture substrate with a reaction mixture, e.g., a reaction mixture comprising e transcriptase, that uses the loaded mRNA as a template, to produce: a first double-stranded cDNA (ds cDNA) comprising a strand that is complementary to a first mRNA that encodes an ACVR from a cell; and a second ds cDNA comprising a strand complementary to a second mRNA encoding a BCVR from the cell (the cDNA loaded capture substrate); wherein the isolated production reaction site, e.g., a production micro-chamber, does not include a nucleic acid encoding a BCVR or an ACVR from a cell other than the cell. d) maintaining the isolated production reaction site, e.g., the production micro-chamber, under conditions that allow amplification of the first and second ds cDNAs, to produce: a plurality of AC ds cDNAs comprising a segment that s an AC t of the ACVR from the cell, e.g., an ACVRS; and a plurality of BC ds cDNAs comprising a segment that encodes a BC element of the BCVR from the cell, e.g., a BCVRS; e) acquiring an isolated linkage reaction site, e.g., a linkage micro-chamber, comprising: covalent linking, e.g., ligation, of a strand of the AC ds cDNA (AC strand) to a strand of the BC ds cDNA (BC strand), wherein the AC and BC strands are both sense strands or antisense strands; and f) amplifying the covalently linked, e.g., d, AC and BC strands. 186. A method of making a y comprising a plurality of unique members, the method comprising: making the plurality of members, n each of the members comprises a sequence that encodes a 0t chain element (AC element) of a 0t chain variable region (ACVR) and a [3 chain element (BC element) of a [3 chain variable region (BCVR), and wherein the ACVR and BCVR are matched, made by a method of any of paragraphs 101-185, wherein each unique nucleic acid sequence of the plurality comprises an AC element and a BC element from a different unique cell, thereby making a library sing a plurality of unique members. 187. The method of paragraph 186, wherein the plurality of unique members ses at least 104, 105, 106, 107, 108, or 109 unique members. 188. The method of paragraph 186 or 187, wherein the plurality of unique members comprises 104 to 109, 104 to 108, 104 to 107, 104 to 106, 104 to 105, 108 to 109, 107 to 109, 106 to 109, 105 to 109, 105 to 108, 106 to 107, 104 to 105, 105 to 106, 106 to 107, 107 to 108, or 108 to 109 unique members. 189. The method of any of paragraphs 186-188, wherein at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%, of the members in the library are unique members (which encode d AC element and BC elements sequences). 190. The method of any of paragraphs 186-189, wherein less than 20%, 15%, 10%, 5%, 4%, 3%, 2%, or 1%, of the s in the library are unique members (which encode matched AC element and BC elements sequences). 191. A library comprising: a plurality of unique members, wherein, i) each unique member of the plurality comprises a segment that encodes an AC element, e.g., an ACVRS, and a segment that encodes a BC element, e.g., a BCVRS, wherein the AC element and the BC t in each unique member is d; ii) each unique member of the plurality comprises a t that encodes an AC element, e.g., an ACVRS, and a segment that s a BC t, e.g., a BCVRS, from a different unique cell; and iii) the library comprises one or more (e.g., two, three, four, or all) of the following properties: a) the library is made by a method of any of paragraphs 101-185; b) the plurality of unique members comprises at least 104, 105, 106, 107, 108, or 109 unique nucleic acid sequences; c) the plurality of unique members comprises 104 to 109, 104 to 108, 104 to 107, 104 to 106, 104 to 105, 108 to 109, 107 to 109, 106 to 109, 105 to 109, 105 to 108, 106 to 107, 104 to 105, 105 to 106, 106 to 107, 107 to 108, or 108 to 109 unique members; d) at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%, of the members in the library are unique members (which encode matched AC element and BC elements sequences); or e) less than 20%, 15%, 10%, 5%, 4%, 3%, 2%, or 1%, of the s in the library are unique members (which encode matched AC element and BC elements sequences). 192. The library of paragraph 191, wherein each unique member of the plurality is configured such that, when expressed, the AC element, e.g., the ACVRS, and the BC element, e.g., the BCVRS, form a functional antigen binding molecule, e.g., a single chain TCR. 193. The library of any of paragraphs 191-192, wherein the y is a y library. 194. The library of any of paragraphs 3, wherein each of the members of the plurality further encodes a polypeptide that results in display of the member on the surface of a display entity. 195. The library of any of paragraphs 191-194, wherein the library is a phage display library. 196. The y of any of aphs 191-194, wherein the library is a yeast display library. 197. The library of any of paragraphs 191-194, wherein the library is a mammalian display library. 198. A method of making a binding polypeptide, the method comprising: a) ing a library of any of paragraphs 191-197; and b) expressing a polypeptide encoded by a unique nucleic acid of the y. 199. The method of paragraph 198, further sing contacting the polypeptide with an antigen. 200. The method of paragraph 198 or 199, further sing retrieving the nucleic acid that s a polypeptide that binds the antigen. 201. A method of making a nucleic acid sequence sing a sequence that encodes an 7 chain element (GC element) of a TCR 7 chain variable region (GCVR) and a 5 chain element (DC element) of a TCR 5 chain variable region (DCVR), and wherein the GCVR and DCVR are matched, the method comprising: a) acquiring an isolated production reaction site, e.g., a production micro-chamber, comprising: i) a 7 chain (GC) strand, wherein the GC strand is a strand of a 7 chain double- stranded cDNA (GC ds cDNA) comprising a t that encodes a GC element of the GCVR from a cell, e.g., a 7 chain variable region ce (GCVRS); and ii) a 5 chain (DC) strand, wherein the DC strand is a strand of a 5 chain double- stranded cDNA (DC ds cDNA) comprising a segment that encodes a DC element of the DCVR from the cell, e.g., a 5 chain variable region sequence (DCVRS), and b) covalent linking, e.g., ligation, of a GC strand to a DC strand, wherein the isolated production reaction site, e.g., a production micro-chamber, does not include a nucleic acid encoding a DCVR or a GCVR from a cell other than the cell, thereby making a nucleic acid ce comprising a sequence that encodes a GC element of a GCVR and a DC element of a DCVR, wherein the GCVR and DCVR are matched. 202. The method of paragraph 201, wherein the GC element comprises, or ts of, a GCVRS, or a functional fragment f (e. g., an antigen binding fragment thereof). 203. The method of paragraph 201 or 202, wherein the DC element ses, or consists of, a DCVRS, or a functional fragment thereof (e.g., an antigen g nt thereof). 204. The method of any of aphs 201-203, wherein the nucleic acid sequence is configured such that, when sed, the GCVRS and the DCVRS form a functional antigen binding molecule, e. 3., a single chain TCR molecule. 205. The method of paragraph 204, wherein the antigen binding molecule is functional in vitro, ex vivo, or in viva, e.g., as determined by a method or assay described herein. 206. The method of any of paragraphs 201-205, wherein acquiring an isolated production reaction site, e.g., a production micro-chamber, comprises: a) acquiring a capture substrate bound to: (i) a first double-stranded cDNA (ds cDNA) comprising a strand that is complementary to a first mRNA that encodes a GCVR from a cell; and (ii) a second ds cDNA sing a strand complementary to a second mRNA encoding a DCVR from the cell (the cDNA loaded capture substrate), and b) maintaining the ed production reaction site, e.g., the production micro-chamber, under conditions that allow amplification of the first and second ds cDNAs, to produce: a plurality of GC ds cDNAs comprising a segment that encodes a GC element of the GCVR from the cell, e. g., a GCVRS; and a plurality of DC ds cDNAs comprising a segment that encodes a DC element of the DCVR from the cell, e.g., a DCVRS. 207. The method of paragraph 206, wherein the GC ds cDNA is identical, or substantially identical, to the first ds cDNA. 208. The method of paragraph 206 or 207, wherein the DC ds cDNA is identical, or substantially identical, to the second ds cDNA. 209. The method of any of paragraphs 206-208, n the capture substrate comprises a bead, e.g., a magnetic bead. 210. The method of any of paragraphs 206-209, wherein the capture substrate comprises a moiety (e.g., an oligonucleotide) which binds to cDNA, e.g., (i) a moiety which binds to the GC ; (ii) a moiety which binds to the DC strand; or (iii) both (i) and (ii). 211. The method of any of paragraphs 0, wherein the first mRNA and the second mRNA are disposed on an mRNA loaded capture substrate. 212. The method of any of paragraphs 206-211, wherein the isolated production reaction site, e.g., the production micro-chamber, comprises: a reagent mixture suitable for producing, from the first and second mRNAs (e.g., after the first and second mRNAs are released from the mRNA loaded capture substrate into a solution), a first ds cDNA sing a segment that encodes a GC element of the GCVR of the cell, e.g., a GCVRS, and a second ds cDNA comprising a segment that encodes a DC element of the DCVR of the cell, e.g., a DCVRS. 213. The method of any of paragraphs 206-212, n the isolated production reaction site, e.g., production micro-chamber, ses primers that mediate the production of the first ds cDNA. 214. The method of any of paragraphs 206-213, n the isolated production reaction site, e.g., tion micro-chamber, comprises primers that mediate the production of the second ds cDNA. 215. The method of any of paragraphs 206-214, wherein a cDNA strand that is complementary to a first mRNA that encodes a GCVR from a cell is made by reverse transcription of the first mRNA. 216. The method of any of paragraphs 206-215, wherein a cDNA strand that is complementary to a second mRNA that encodes a DCVR from a cell is made by reverse transcription of the second mRNA. 217. The method of paragraph 215 or 216, wherein the reverse transcription takes place in the isolated production reaction site, e.g., a production-micro r. 218. The method of paragraph 215 or 216, wherein the reverse transcription takes place in an isolated cell reaction site, e. g., a cell isolation micro-chamber. 219. The method of paragraph 215 or 216, wherein the reverse transcription takes place outside the isolated production reaction site, e.g., a production micro-chamber, or outside an isolated cell reaction site, e.g., a cell ion micro-chamber. 220. The method of paragraph 215 or 216, wherein the reverse transcription takes place outside the isolated production reaction site, e.g., a production-micro chamber, and outside an isolated cell reaction site, e.g., a cell isolation micro-chamber. 221. The method of paragraph 215 or 216, wherein the reverse transcription takes place outside an isolated reaction site, e.g., e a micro-chamber. 222. The method of any of paragraphs 206-221, wherein the amplification comprises 20 or fewer cycles, e.g., 25 or fewer, 24 or fewer, 23 or fewer, 22 or fewer, 21 or fewer, 20 or fewer, 9 or fewer, 8 or fewer, 7 or fewer, 6 or fewer, or 5 or fewer cycles. 223. The method of any of paragraphs 206-222, n the reverse transcription and/or amplification uses one or more primers, e.g., sing a sequence c for a GCVRS and/or a DCVRS. 224. The method of any of paragraphs 206-223, wherein the amplification ses using two or more primers that mediate the production of the GC ds cDNA, wherein at least one primer comprises a nucleotide modification, and wherein at least one primer does not comprise a tide modification. 225 . The method of any of paragraphs 206-224, wherein the amplification comprises using two or more primers that mediate the production of the DC ds cDNA, wherein at least one primer comprises a nucleotide modification, and wherein at least one primer does not comprise a nucleotide modification. 226. The method of paragraph 225, wherein at least one primer comprises a nucleotide modification, e.g., which reduces, e.g., inhibits, DNA synthesis, e.g., by a DNA polymerase. 227. The method of paragraph 225 or 226, wherein at least one primer does not comprise a nucleotide modification, e.g., which reduces, e.g., inhibits, DNA synthesis, e.g., by a DNA polymerase. 228. The method of paragraph 226 or 227, wherein the nucleotide modification inhibits a DNA rase from extending the DNA. 229. The method of any of paragraphs 226-228, wherein the nucleotide modification is an insertion of a spacer to the primer, e.g., between two adjacent nucleotides in the primer. 230. The method of paragraph 229, wherein the spacer is a flexible spacer, a carbon spacer (e.g., n-, wherein n=3, 4, 5, or more), two or more (6.3., three, four, five, or more) abasic tides or a PEG spacer. 231. The method of any of paragraphs 226-228, wherein the nucleotide modification is 2’-O- methyl, 2’-OH, 2’-NH2, or uracil, e.g., to a ribose. 232. The method of any of paragraphs 226-231, wherein the nucleotide modification is d ally or at the 3’ end of the primer. 233. The method of any of paragraphs 223-232, wherein at least one primer ses (i) a first member; (ii) a second ; and optionally (iii) a nucleotide modification described herein, e.g., located between (i) and (ii). 234. The method of paragraph 233, wherein the first member is capable of annealing with the second member in the same primer or a different primer, e.g., forming a hairpin ure (Via intramolecular hybridization) or a double-stranded ure (Via intermolecular hybridization) comprising a duplex region of 4, 5, 6, 7, 8, 9, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 20, more basepairs. 235 . The method of paragraph 233 or 234, n the first member comprises a sequence that is mentary to the sequence of an oligonucleotide attached to the capture substrate. 236. The method of any of paragraphs 233-235, wherein the second member comprises (6g, from 5 ’ to 3’) one, two, or all of: (i) a ce that is complementary to at least a portion of the first member; (ii) a universal priming sequence (e.g., for PCR ication or next-generation sequencing); and (iii) a sequence complementary to a target sequence, e.g., a GCVRS and/or a DCVRS, optionally, wherein the second member comprises a sequence for homologous recombination (e.g., in a yeast or mammalian cell). 237. The method of any of paragraphs 223-236, wherein at least one primer comprises a sequence encoding at least a n of a linker sequence, or a complementary sequence thereof. 238. The method of paragraph 237, n the primer that comprises a sequence encoding at least a portion of a linker sequence, or a complementary sequence thereof, is phosphorylated, e.g., ’ phosphorylated, 239. The method of paragraph 237 or 238, wherein the linker sequence comprises, or consists of, (Gly-Gly-Gly-Gly-Ser)n, where n=1, 2, 3, 4, 5, or more. 240. The method of any of paragraphs 201-239, n the GC ds cDNA ses a 5’ overhang, e.g., a 5’ overhang that is capable of izing to an oligonucleotide attached to a capture substrate. 241. The method of any of paragraphs 201-240, wherein the GC ds cDNA comprises a blunt end, e.g., a blunt end comprising a 5’ phosphate. 242. The method of any of paragraphs 201-241, wherein the DC ds cDNA comprises a 5’ overhang, e.g., a 5’ overhang that is capable of hybridizing to an oligonucleotide attached to a capture substrate. 243. The method of any of paragraphs 2, wherein the DC ds cDNA comprises a blunt end, e.g., a blunt end comprising a 5’ phosphate. 244. The method of any of paragraphs 201-243, wherein the GC ds cDNA and the DC ds cDNA comprise sticky ends, e.g., both have 5’ overhangs. 245. The method of any of paragraphs 201-244, wherein the GC strand and the DC strand are covalently linked, e.g., ligated, to produce a single stranded nucleic acid ce, wherein the GC and DC strands are both sense strands or both antisense strands. 246. The method of any of paragraphs 201-245, wherein a denatured GC strand of the GC ds cDNA to a denatured DC strand of the DC ds cDNA are covalently linked, e.g., ligated, n the GC and DC strands are both sense s or both antisense strands. 247. The method of any of paragraphs 201-245, wherein the GC strand is present in the GC ds cDNA and the DC strand is present in the DC ds cDNA, and wherein the GC ds cDNA and the DC ds cDNA are covalently linked, e.g., ligated, e.g., to produce a double stranded nucleic acid sequence. 248. The method of any of paragraphs 7, wherein the covalent linking, e. g., ligation, occurs in the isolated production reaction site. 249. The method of paragraph 248, wherein the ed production reaction site, e.g., a production micro-chamber, comprises a reagent that is capable of covalently linking, e.g., ligating, the GC and DC strands or the GC and DC ds cDNAs. 250. The method of paragraph 248 or 249, wherein the isolated production reaction site, e.g., a production micro-chamber comprises an enzyme that ntly couples the GC and DC strands or the GC and DC ds cDNAs. 251. The method of any of paragraphs 201-247, wherein the covalent g, e.g., ligation, occurs in a site different from the isolated production reaction site, e.g., occurs in an ed linkage reaction site, e.g., a linkage micro-chamber. 252. The method of paragraph 251, wherein the GC strand and the DC strand are transferred from the isolated production site to the ed linkage reaction site, e. g., a linkage micro-chamber, and the covalent linking occurs in the isolated linkage reaction site, e.g., a linkage micro-chamber. 253. The method of paragraph 251 or 252, wherein the isolated linkage reaction site, e.g., a linkage micro-chamber, comprises a reagent that is capable of covalently linking, e.g., ligating, the GC and DC strands or the GC and DC ds cDNAs. 254. The method of any of paragraphs 25 1-25 3, wherein the isolated linkage reaction site, e.g., a e micro-chamber, comprises an enzyme that covalently s the GC and DC strands or the GC and DC ds cDNAs. 255. The method of paragraph 250 or 254, wherein the enzyme is a ligase, e.g., a thermal stable ligase. 256. The method of any of paragraphs 251-255, wherein the covalent linking, e.g., ligation, comprises: (a) heating the isolated linkage reaction site, e.g., the linkage micro-chamber, under conditions (e.g., at 96°C) that allow ration of the GC strand and the DC strand; (b) cooling the isolated linkage reaction site, e.g., the linkage micro-chamber, under conditions (e.g., at 60-66°C) that allow hybridization of the splint oligonucleotide to the GC strand and the DC ; (c) maintaining the isolated linkage reaction site, e.g., the linkage chamber, under conditions (e.g., at 46-66°C) that allow ligation of the GC strand and the DC strand (e.g., formation of phosphodiester bond between the GC strand and the DC strand); and (d) repeating steps (a), (b), and (c) sequentially for 2, 6, 20, 26, 20, 26, 30, 36, 40, 46, 60, or more cycles. 257. The method of any of aphs 201-256, wherein the GC strand and the DC strand are covalently linked, e.g., ligated, in the presence of a splint oligonucleotide. 258. The method of paragraph 257, wherein the splint oligonucleotide is ized to a sequence comprising the on of the GC strand and the DC strand, or a ce complementary thereof, and forms a duplexed region at the site of on. 259. The method of aph 257 or 258, wherein the splint oligonucleotide comprises a modification (e.g., an NHZ group) that inhibits DNA synthesis, e.g., by a DNA polymerase. 260. The method of paragraph 259, wherein the modification is at the 3’ end of the splint oligonucleotide. 261. The method of any of paragraphs 201-260, wherein a strand complimentary to the covalently linked, e. g., ligated, GC and DC s is produced by amplification. 262. The method of any of paragraphs 201-261, further comprising, prior to acquiring the isolated production reaction site, e.g., a production micro-chamber, acquiring an mRNA loaded capture substrate. 263. The method of paragraph 262, wherein acquiring the mRNA loaded capture substrate comprising: a) acquiring an ed cell reaction site, e.g., a cell ion micro-chamber, comprising: i) a cell; and ii) a capture substrate capable of binding a first mRNA encoding a GCVR from the cell and a second mRNA encoding a DCVR from the cell; and b) maintaining the ed cell reaction site, e.g., the cell isolation micro-chamber, under conditions that allow lysis of the cell and binding of the capture substrate with the first mRNA and the second mRNA to form the mRNA loaded capture substrate, wherein the isolated cell on site, e.g., cell isolation micro-chamber, does not e a nucleic acid encoding a GCVR or a DCVR from a cell other than the cell. 264. The method of paragraph 263, wherein the isolated cell on site, e.g., cell isolation micro-chamber, comprises a lysing reagent, e.g., a detergent. 265. The method of paragraph 263 or 264, n the cell is lysed by heat or an enzyme. 266. The method of any of paragraphs 263-265, wherein the capture substrate ses a moiety (e.g., an oligonucleotide) which binds mRNA, e.g., an oligo(dT). 267. The method of any of paragraphs 262-266, further comprising releasing the mRNA loaded capture substrate from the isolated cell reaction site, e.g., the cell isolation micro-chamber. 268. The method of aph 267, wherein the releasing step is performed in the presence of a poly(dA) or poly(dT) ucleotide, e.g., to reduce cross-binding of ptured mRNA. 269. The method of paragraph 262-268, wherein the mRNA loaded capture substrate is transferred from the isolated cell reaction site, e.g., the cell isolation micro-chamber, to the isolated production reaction site, e.g., the production micro-chamber. 270. The method of any of paragraphs 201-269, comprising releasing the nucleic acid sequence from the production micro-chamber. 271. The method of any of paragraphs 201-270, further comprising amplifying the nucleic acid sequence. 272. The method of any of paragraphs 201-271, comprising sequencing all or a portion of the nucleic acid sequence. 273. The method of any of aphs 201-272, comprising inserting all or a portion of nucleic acid sequence into a vector. 274. The method of paragraph 273, wherein the vector supplies an onal GC element or DC element not included in the nucleic acid sequence. 275. The method of paragraph 273 or 274, n the vector supplies a GC CDRl, a GC CDR2, or both. 276. The method of any of aphs 273-275, comprising expressing the vector. 277. The method of any of paragraphs 201-276, comprising expressing the nucleic acid sequence to produce a ptide comprising a segment that encodes a GC element of the GCVR, e.g., a GCVRS, and a segment that encodes a DC element of the DCVR, e.g., a DCVRS. 278. The method of paragraph 277, wherein the DC element is N—terminal to the GC element in the ptide. 279. The method of paragraph 277, wherein the GC element is C-terminal to the DC element in the polypeptide. 280. The method of any of paragraphs 277-279, r comprising contacting the polypeptide with an antigen. 281. The method of any of paragraphs 0, r comprising determining if the polypeptide binds the antigen. 282. The method of any of paragraphs 201-281, wherein the cell is an immune cell, e.g., a B cell or T cell, e.g., a human B cell or T cell. 283. The method of any of paragraphs 201-282, wherein the cell is a mammalian cell or an avian cell. 284. A method of making a nucleic acid sequence comprising a sequence that encodes a 7 chain element (GC element) of a TCR y chain variable region (GCVR) and a 5 chain element (DC element) of a TCR 5 chain variable region (DCVR), and n the GCVR and DCVR are matched, comprising: a) acquiring an isolated cell on site, e.g., a cell isolation micro-chamber, comprising: i) a cell; and ii) a capture ate capable of binding a first mRNA encoding a GCVR from the cell and a second mRNA encoding a DCVR from the cell; b) maintaining isolated cell reaction site, e.g., the cell isolation micro-chamber, under conditions that allow lysis of the cell and binding of the capture substrate with the first mRNA and the second mRNA to form the mRNA loaded capture substrate, n the isolated cell reaction site, e.g., cell isolation micro-chamber, does not include a nucleic acid encoding a GCVR or a DCVR from a cell other than the cell; c) contacting the mRNA loaded capture substrate with a reaction mixture, e.g., a reaction mixture comprising reverse transcriptase, that uses the loaded mRNA as a template to make cDNA (this can occur, e.g., in the ed cell reaction site, in the isolated production reaction site, or in neither, e. g., not in an ed reaction site); d) acquiring an isolated production reaction site, e.g., a production micro-chamber, comprising: WO 19402 i) an y chain (GC) strand from step b), wherein the GC strand is a strand of a 7 chain double-stranded cDNA (GC ds cDNA) comprising a segment that encodes a GC element of the GCVR from the cell, e.g., a 7 chain variable region sequence ); and ii) a 5 chain (DC) strand from step b), wherein the DC strand is a strand of a 5 chain double-stranded cDNA (DC ds cDNA) comprising a segment that encodes a DC element of the DCVR from the cell, e.g., a 5 chain variable region sequence (DCVRS); and e) covalent linking, e. g., ligation, of a GC strand to a DC , wherein the isolated production reaction site, e.g., a production micro-chamber, does not include a nucleic acid encoding a DCVR or a GCVR from a cell other than the cell. 285. A method of making a nucleic acid sequence comprising a sequence that encodes a y chain element (GC element) of a TCR 7 chain variable region (GCVR) and a 5 chain element (DC element) of a TCR 5 chain variable region (DCVR), and wherein the GCVR and DCVR are matched, comprising: a) acquiring an ed cell reaction site, e.g., a cell isolation micro-chamber, comprising: i) a cell; and ii) a capture substrate e of binding a first mRNA ng a GCVR from the cell and a second mRNA encoding a DCVR from the cell; b) maintaining the isolated cell reaction site, e.g., the cell isolation micro-chamber, under conditions that allow lysis of the cell and binding of the capture substrate with the first mRNA and the second mRNA to form the mRNA loaded capture substrate, wherein the isolated cell reaction site, e.g., cell ion micro-chamber, does not include a nucleic acid encoding a GCVR or a DCVR from a cell other than the cell; c) ing an ed production reaction site, e.g., a production micro-chamber, comprises: contacting the mRNA loaded capture substrate with a reaction e, e.g., a reaction mixture comprising reverse transcriptase, that uses the loaded mRNA as a template, to produce: a first double-stranded cDNA (ds cDNA) comprising a strand that is complementary to a first mRNA that encodes a GCVR from a cell; and a second ds cDNA comprising a strand complementary to a second mRNA encoding a DCVR from the cell (the cDNA loaded e substrate); wherein the isolated production on site, e.g., a production micro-chamber, does not include a nucleic acid encoding a DCVR or a GCVR from a cell other than the cell. d) maintaining the isolated production on site, e.g., the production micro-chamber, under conditions that allow amplification of the first and second ds cDNAs, to produce: a plurality of GC ds cDNAs comprising a segment that encodes a GC element of the GCVR from the cell, e. g., a GCVRS; and a plurality of DC ds cDNAs comprising a segment that encodes a DC element of the DCVR from the cell, e.g., a DCVRS; e) acquiring an ed linkage reaction site, e.g., a linkage micro-chamber, comprising: covalent linking, e.g., ligation, of a strand of the GC ds cDNA (GC strand) to a strand of the DC ds cDNA (DC ), n the GC and DC strands are both sense strands or antisense strands; and f) amplifying the covalently linked, e.g., ligated, GC and DC s. 286. A method of making a library comprising a plurality of unique members, the method comprising: making the plurality of members, wherein each of the members comprises a sequence that encodes a 7 chain element (GC element) of a 7 chain variable region (GCVR) and a 5 chain element (DC element) of a 5 chain variable region (DCVR), and wherein the GCVR and DCVR are matched, made by a method of any of paragraphs 201-285, wherein each unique nucleic acid sequence of the plurality comprises a GC element and a DC element from a different unique cell, thereby making a library comprising a plurality of unique members. 287. The method of paragraph 86, wherein the plurality of unique s comprises at least 204, 205, 206, 207, 208, or 209 unique members. 288. The method of paragraph 286 or 287, wherein the plurality of unique members comprises 204 to 209, 204 to 208, 204 to 207, 204 to 206, 204 to 205, 208 to 209, 207 to 209, 206 to 209, 205 to 209, 205 to 208, 206 to 207, 204 to 205, 205 to 206, 206 to 207, 207 to 208, or 208 to 209 unique 289. The method of any of paragraphs 286-288, wherein at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 200%, of the members in the library are unique members (which encode d GC element and DC elements ces). 290. The method of any of paragraphs 286-289, wherein less than 20%, 25%, 20%, 5%, 4%, 3%, 2%, or 1%, of the members in the library are unique members (which encode matched GC element and DC elements sequences). 291. A library comprising: a plurality of unique members, wherein, i) each unique member of the plurality comprises a segment that encodes a GC element, e.g., a GCVRS, and a segment that encodes a DC element, e.g., a DCVRS, wherein the GC element and the DC element in each unique member is matched; ii) each unique member of the ity comprises a segment that s a GC element, e. g., a GCVRS, and a segment that encodes a DC element, e.g., a DCVRS, from a different unique cell; iii) the library comprises one or more (e.g., two, three, four, or all) of the ing properties: a) the library is made by a method of any of paragraphs 201-285; b) the plurality of unique s comprises at least 204, 205, 206, 207, 208, or 209 unique nucleic acid sequences; c) the plurality of unique members comprises 204 to 209, 204 to 208, 204 to 207, 204 to 206, 204 to 205, 208 to 209, 207 to 209, 206 to 209, 205 to 209, 205 to 208, 206 to 207, 204 to 205, 205 to 206, 206 to 207, 207 to 208, or 208 to 209 unique s; d) at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 200%, of the members in the library are unique members (which encode matched GC element and DC elements sequences); or e) less than 20%, 25%, 20%, 5%, 4%, 3%, 2%, or 1%, of the members in the library are unique s (which encode matched GC element and DC elements sequences). 292. The library of paragraph 291, wherein each unique member of the ity is configured such that, when expressed, the GC element, e.g., the GCVRS, and the DC element, e.g., the DCVRS, form a functional antigen binding molecule, e.g., a single chain TCR. 293. The library of any of paragraphs 291-292, wherein the library is a display library. 294. The library of any of paragraphs 291-293, wherein each of the members of the plurality further encodes a polypeptide that s in display of the member on the surface of a display entity. 295. The library of any of paragraphs 291-294, wherein the library is a phage display library. 296. The library of any of paragraphs 4, wherein the library is a yeast display library. 297. The library of any of paragraphs 291-294, n the library is a mammalian display library. 298. A method of making a binding polypeptide, the method comprising: a) acquiring a library of any of paragraphs 291-297; and b) expressing a polypeptide encoded by a unique nucleic acid of the library. 299. The method of paragraph 298, further comprising contacting the polypeptide with an antigen. 300. The method of paragraph 298 or 299, further comprising retrieving the nucleic acid that encodes a ptide that binds the antigen. 301. An isolated tion on site, e.g., a production micro-chamber, which is an isolated production reaction site described in any of paragraphs l-85, 101-185, or 5. 302. The isolated production reaction site, e.g., a production micro-chamber, of aph 401, which does not include a nucleic acid ng (i) an HCVR or an LCVR, (ii) an ACVR or a BCVR, or (iii) a GCVR or a DCVR, from a cell other than the cell, 303. The isolated production reaction site, e.g., a production micro-chamber, of paragraph 301 or 302, which comprises one, two, or all of: (i) one or more primers specific to V gene sequences of the (a) HC and LC, (b) a chain and [3 chain, or (c) 7 chain and 5 chain; (ii) one or more primers specific to overhangs introduced onto the (a) HC and LC, (b) or chain and [3 chain, or (c) 7 chain and 6 chain, cDNAs; (iii) one or more primers comprising a first member, a second member, and a nucleotide modification (e.g., a spacer) located between the first and second members, wherein the first member is capable of annealing with the second member in the same primer or a ent primer, e. g., forming a hairpin ure (via intramolecular hybridization) or a double-stranded structure (via intermolecular hybridization) comprising a duplex region of 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, more basepairs. 304. The isolated production reaction site, e.g., a production micro-chamber, of any of paragraphs 301-103, which does not comprise a reagent that can covalently link nucleic acids, e.g., a ligase, e.g., a thermostable ligase. 305. The isolated production reaction site, e.g., a production micro-chamber, of any of aphs 301-303, which comprises a reagent that can ntly link nucleic acids, e.g., a ligase, e. g., a thermostable ligase. 306. A self-annealing oligonucleotide comprising a first member, a second member, and a nucleotide modification (e.g., a ) located between the first and second members, wherein the first member is capable of annealing with the second member in the same oligonucleotide or a different oligonucleotide, e. g., forming a hairpin structure (via intramolecular hybridization) or a -stranded structure (via intermolecular hybridization) comprising a duplex region of 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, more basepairs. 307. The ucleotide of paragraph 306, wherein the first and second s are capable of forming a hairpin structure comprising a duplex region of 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, , 16, 17, 18, 19,20, more basepairs. 308. The oligonucleotide of paragraph 306 or 307, wherein the first member is 5-40 nucleotides, e.g., 5-10, 5-20, 5-30, 30-40, 20-40, 10-30, 10-30, or 15-25 nucleotides, in length. 309. The oligonucleotide of any of paragraphs 306-308, wherein the second member is 5-40 nucleotides, e.g., 5-10, 5-20, 5-30, 30-40, 20-40, 10-30, 10-30, or 15-25 nucleotides, in . 310. The oligonucleotide of any of paragraphs 306-309, n the spacer is a flexible spacer or a PEG spacer. 311. The oligonucleotide of any of paragraphs 0, wherein the first member comprises a sequence that is complementary to the sequence of an oligonucleotide attached to a e substrate. 312. The ucleotide of any of paragraphs 306-311, wherein the second member comprises (e.g., from 5’ to 3’) one, two, or all of: (i) a sequence that is complementary to at least a portion of the first member; (ii) a universal priming sequence (e.g., for PCR amplification or next- generation sequencing); and (iii) a sequence mentary to a target sequence, e.g., a GCVRS andfor a DCVRS, ally, wherein the second member comprises a sequence for homologous recombination (e.g., in a yeast or ian cell). 313. An isolated linkage reaction site, e. g., a linkage micro-chamber, which is an isolated linkage reaction site described in any of paragraphs 51-83, 85, 151-183, 185, 251-283, or 285. 314. The ed linkage reaction site, e.g., a linkage micro-chamber, of paragraph 313, which does not include a nucleic acid encoding (i) an HCVR or an LCVR, (ii) an ACVR or a BCVR, or (iii) a GCVR or a DCVR, from a cell other than the cell, 315. The isolated linkage reaction site, e.g., a linkage micro-chamber, of paragraph 313 or 314, which comprises a splint oligonucleotide that is capable of hybridizing to a sequence comprising the junction of (i) the HC strand and the LC strand, (ii) the AC strand and the BC strand, or (iii) the GC strand and the DC strand, or a sequence complementary thereof, to form a duplexed region at the site of ligation. 316. The isolated linkage reaction site, e.g., a linkage micro-chamber, of any of paragraphs 313-315, which ses a reagent that can covalently link nucleic acids, e.g., a , e.g., a thermostable ligase. 317. The method of any of paragraphs 1-300, which does not include a step of overlap extension polymerase chain reaction (OE-PCR), also known as splicing by overlap extension or splicing by overhang extension (SOE) PCR (Higuchi er al., Nucleic Acids Res. 1988; 16(15):7351- 67), e.g., in the linking step.
EXAMPLES Example 1: Cohesive-End PCR-Ligation in Drops B cells were encapsulated in ts. B cells were encapsulated into droplets, with droplet volume ranging from 10 pL to 100 nL, typically 000 pL. s of B cells can e, for example, mice, human, rat, rabbit, or chicken. The carrier (oil) phase was composed of 3M HFE- 7500 with ~1% urfactant (RAN Biotechnologies). Droplets were formed by a microfluidic chip (e.g., 2R 100 pm from Dolomite) with flow of fluid phases controlled by a syringe or pressure pump. The aqueous phase of droplets was composed of a buffer (e.g., Tris, pH 7.5), a detergent to aid cell lysis, and magnetic beads which contain oligonucleotides (primers) to anneal to heavy and light chain mRNAs. Occupancy of drops should be not more than 1 cell per droplet, and at least one bead per droplet.
The droplets were incubated to facilitate cell lysis. The on was heated to improved lysis efficiency in the ce of certain detergents (e.g., Tween 20). For e, the emulsion can be heated to a temperature of 40°C, 50°C, 60°C, 70°C, or 80°C, for approximately 5-60 minutes.
After the cells were lysed, mRNA was released and captured on beads by annealing to oligonucleotides on the beads.
The on was broken and the beads were red. Emulsions (or coalesce different solution phases) were broken using drop destabilizing reagent, such as PFO (perfluorooctanol). The aqueous phase containing the beads was recovered. The beads were isolated using magnet. The beads were washed and resuspended in a buffer (e.g., Tris, pH 7.5).
Reverse transcription (RT) was performed to create cDNA-beads (in a non-emulsion reaction). The beads were resuspended in a buffer-enzyme mix to facilitate RT (e.g., Superscript II RT). The reaction was incubated at 40°C for 15 minutes to facilitate RT. The beads were washed once with a buffer (6.3., Tris, pH 7.5).
The recovered beads can be ulated for PCR-Ligation reaction. Droplets g in volume from about 5 pL-500 pL, most commonly about 10-50 pL, can be used. The carrier (oil) phase can be composed of 3M HFE-7500 containing ~2% fluorosurfactant (RAN hnologies).
Drops can be encapsulated with: beads which have cDNA; PCR reagents (including, e.g., a DNA polymerase, e.g., Phusion® High-Fidelity DNA Polymerase (NEB), Q5® High-Fidelity DNA Polymerase (NEB), Pfu DNA rase, KAPA DNA polymerase, Vent® DNA polymerase, or Taq DNA polymerase); ucleotides that allow for amplification of VH and VL sequences; a thermostable ligase (e.g., Taq DNA ligase, Pfu DNA ligase, Ampligase® thermostable DNA ligase, Tsc DNA ligase, Rma DNA ligase, Tfi DNA ligase, or Tth DNA ligase); and optimized buffer conditions (compatibility to support both DNA polymerase and ligase enzymatic activities).
The scFV cassette was constructed as VL-Linker-VH, as tested in tubes. The order can be switched (VH-Linker-VL) with no cant impact on function or performance. The e s of the VL sequence can contain an overhang sequence encoding a Linker sequence and with at least 1 modified nucleotide (e.g., 3 consecutive nucleotides with 2’-O-methyl modification). The VL reverse primers can also contain 5’-phosphate (required to be substrate of ligase). The forward primers of the VH sequence can contain an overhang sequence encoding a Linker sequence and with at least 1 modified nucleotide (e.g., 3 consecutive nucleotides with 2’-O-methyl modification). The VH forward primers can also contain 5’-phosphate red to be substrate of ligase). The occupancy of drops should be not more than 1 bead per drop.
Thermocycling can be med with emulsion. The ted emulsion can be transferred to PCR tubes. Thermocycling can be performed using the following conditions: initial denaturation at 95-98°C for 30 s to 2 s; 10-30 cycles of: denaturation at 95-98°C for 10-30 seconds, primer annealing at 50-60°C for 10-30 seconds, polymerase extension at 72°C for 30 seconds, and ve product annealing and ligation at 45-55°C for 3 minutes. The reaction can be hold at 4°C.
The emulsion was broken and the portion which contains linked (and non-linked) t was recovered. Emulsions can be broken using drop destabilizing reagent, such as PFO (perfluorooctanol). The s phase can be recovered and the beads can be discarded.
The linked product (representing natively linked VL—linker-VH), as tested in tubes, was purified from non-linked VH and VL products. Ligated product was separated from non-ligated products by size separation. For example, denaturing PAGE (polyacrylamide gel electrophoresis) or denaturing HPLC-SEC can be used. Linked product (about 800-900 bp) was isolated from non-linked product (about 350-500 bp). For denaturing PAGE purification, the ligated band was cut out from the gel and electroelution was performed to extract DNA from the gel slice (Bio-Rad Electro-Elutor).
The purified linked product can be amplified by PCR. Polymerase/conditions which can moderately read through DNA ning modified nucleotides, e.g., Taq polymerase, can be used.
Final PCR product can be introduced to yeast using standard methods (e.g., oporation with expression vector) to create a natively paired library derived from biological sources.
Example 2: Ligase Cycling Approach B cells were encapsulated in droplets. B cells were encapsulated into droplets, with droplet volume ranging from 10 pL to 100 nL, typically 000 pL. Sources of B cells can include, for example, mice, human, rat, rabbit, or chicken. The carrier (oil) phase was composed of 3M HFE- 7500 with ~1% fluorosurfactant (RAN Biotechnologies). Droplets were formed by a microfluidic chip (e.g., 2R 100 pm from Dolomite) with flow of fluid phases controlled by a syringe or pressure pump. The aqueous phase of droplets was composed of a buffer (e.g., Tris, pH 7.5), a detergent to aid cell lysis, and magnetic beads which contain ucleotides (primers) to anneal to heavy and light chain mRNAs. Occupancy of drops should be not more than 1 cell per droplet, and at least one bead per droplet.
The droplets were incubated to facilitate cell lysis. The on was heated to improved lysis efficiency in the presence of certain detergents (e.g., Tween 20). For example, the emulsion can be heated to a temperature of 40°C, 50°C, 60°C, 70°C, or 80°C, for approximately 5-60 minutes.
After the cells were lysed, mRNA was released and captured on beads by annealing to ucleotides on the beads.
The emulsion was broken and the beads were recovered. Emulsions (or coalesce different solution phases) were broken using drop destabilizing reagent, such as PFO (perfluorooctanol). The aqueous phase ning the beads was recovered. The beads were isolated using magnet. The beads were washed and resuspended in a buffer (6.3., Tris, pH 7.5).
Reverse transcription (RT) can be performed to create eads (in a non-emulsion reaction). The beads can be resuspended in a -enzyme mix to facilitate RT (e.g., Superscript 11 RT). The reaction can be ted at 40°C for 15 minutes to facilitate RT. The beads can be washed once with a buffer (e.g., Tris, pH 7.5).
Recovered beads were encapsulated for PCR reaction. Droplets ranging in volume from about 5 pL—500 pL, most commonly about 10-50 pL, can be used. The carrier (oil) phase can be composed of 3M 00 ning ~2% fluorosurfactant (RAN Biotechnologies). Drops can be encapsulated with: beads which contain cDNA (some beads can be ‘empty’, which is not problematic); PCR ts (including, e.g., a DNA polymerase, e.g., n® High-Fidelity DNA rase (NEB), Q5® idelity DNA Polymerase (NEB), Pfu DNA rase, KAPA DNA polymerase, Vent® DNA polymerase, or Taq DNA polymerase); oligonucleotides that allow for amplification of VH and VL sequences.
The scFv cassette can be constructed as VL—Linker-VH. The order can be switched (VH- Linker-VL) with no significant impact on function or performance. The VL reverse and VH d primers can contain an overhang sequence encoding Linker. The VH e primer can have a 5’- phosphate. The occupancy of drops should be not more than 1 bead per drop.
Thermocycling can be performed with on. The generated emulsion can be erred to PCR tubes. The thermocycling can be performed using standard PCR conditions: initial denaturation: 95-98°C for 30 seconds to 2 minutes; 10-30 cycles of: denaturation at 95-98°C for 10- seconds, primer annealing at 50-60°C for 10-30 seconds, polymerase extension at 72°C for 30 seconds. The reaction can be hold at 4°C.
The on can be broken and the beads can be recovered. Emulsions (or coalesce different solution phases) can be broken using drop destabilizing reagent, such as PFO (perfluorooctanol). The aqueous phase that contains beads can be recovered. The beads can be isolated using magnet. The beads can be washed to remove PCR product not captured on beads. The beads can be resuspended in a buffer (e.g., Tris, pH 7.5).
The recovered beads can be ulated for ligase cycling reaction. Droplets g in volume from about 5 pL-SOO pL, most ly about 10-50 pL, can be used. The carrier (oil) phase can be composed of 3M HFE-7500 containing ~2% fiuorosurfactant (RAN Biotechnologies).
The drops can be encapsulated with a Stint oligonucleotide which is complementary and anneals to 3’ terminus of ‘top’ VL strain and 5’ terminus of ‘top’ VH strand; a thermostable ligase (e.g., Taq DNA ligase, Pfu DNA ligase, Ampligase® thermostable DNA ligase, Tsc DNA ligase, Rma DNA ligase, Tfi DNA ligase, or Tth DNA ligase); reaction ents ed to support ligase enzymatic activity (e.g., NAD). The occupancy should be not more than 1 bead per drop.
Thermocycling can be performed with emulsion. The ted emulsion can be transferred to PCR tubes. Thermocycling can be performed using rd conditions: 3-15 cycles of: denaturation: 90-95°C for 30 seconds, annealing and ligation at 50-60°C for 1-3 minutes. The reaction can be hold at 4°C.
The emulsion can be broken and the aqueous portion which contains the linked product can be recovered. Emulsions (or coalesce different solution phases) can be broken using drop destabilizing reagent, such as PFO (perfluorooctanol). The aqueous phase can be recovered and the beads can be discarded.
The purified linked product can be amplified by PCR. Standard conditions can be used with oligonucleotides that anneal the outer termini of the linked VL-Linker-VH d fragment.
Final PCR product can be introduced to yeast to create a natively paired library derived from biological sources.
Example 3: Ligase Cycling Reaction In this example, VH and VL PCR products were covalently linked using a thermostable ligase and a splint oligo.
RNA from a hybridoma clone (ATCC, HB-112, “4G2”) was isolated using an RNeasy Kit n). cDNA was generated using SuperScript IV Reverse Transcriptase (Thermo Fisher) and the isolated RNA ing manufacturer recommended conditions. Primers used were directed to nt regions of the heavy and light chains: mouse IgG_CHl_rev (5’- CHGATGGGGSTGTYGTTKTRGC (SEQ ID NO: 1)) and mouse Kappa_Rev (5 ’- GTGCAGCATCAGCCC (SEQ ID NO: 2)). As used herein, the nucleotides are defined by IUPAC nucleotide code, e.g., R=A or G; Y=C or T; S: G or C; K=G or T; H=A or C or T.
The components were mixed, heated to 65 °C for 5 min, and then incubated on ice for at least 1 min. The following components were then added: Component Volume (111) 5X SSIV buffer 4 100mm RNaseOUT RNase Inhibitor (40 U/ul) SuperScript IV Reverse Transcriptase The components were mixed and then incubated at 55°C for 10 minutes. The reaction was then inactivated by incubating at 80°C for 10 minutes. cDNA ts were used as templates for PCR to separately generate VH and VL products. The s used ed: 4G2 VL_Fwd 5’-GACATCAAGATGACCCAGTCTC (SEQ ID NO: 3) Mouse Kappa_Rev-Phos ’-/5Phos/ACCAGCAGAGCTCTCACCTGGTGCAGCATCAGCCC (SEQ ID NO: 4) 4G2 VH_Fwd-Phos ’-/5Phos/GGAACTACCGAAGGCACAGGTGAGGTCCAGCTGCAACAGTC (SEQ ID NO: 5) Mouse IgG_CHl_rev ’-CHGATGGGGSTGTYGTTKTRGC (SEQ ID NO: 1) WO 19402 The PCR reactions each included: Q5 Hot Start 2X Master Mix (NEB) Thermocycling was then performed as follows: Initial Denature Anneal Extend Denaturation Forever PCR products were purified using AMPure beads. Purified PCR ts were quantified by op. Ligase cycling reactions were set up in 25 ul reactions to achieve a 1:1:1 molar ratio of VH product, VL product, and splint oligo. The splint oligo used had the following sequence: ’—ACCTGTGCCTTCGGTAGTTCCACCAGCAGAGCTCTCACCTG/3AmMO/ (SEQ ID NO: 6).
Each ligase cycling reaction included: Component Amount 4G2 VH PCR product 38 ng 4G2 VL PCR t 35 ng Splint oligo (0.1 mM) 3.3 ul Taq ligase buffer (NEB) 2.5 ul Taq ligase (NEB) 1 ul Water Up to 25 ul The ligase cycling reactions were analyzed by denaturing PAGE (polyacrylamide gel electrophoresis). In addition to Taq Ligase, an onal thermostable ligase (Ampligase (Amp), Lucigen) was evaluated.
As shown in efficient linking of VH and VL product was observed using both Taq ligase and Ampligase thermostable ligase.
Example 4: Retention of Native g During Bulk Re-Amplification of Linked Products An emulsion OE-PCR workflow creates VH and VL products which arily share ces to facilitate splicing by p extension. In such a w, any retained VH/VL product which is not spliced together within a drop can become spliced during bulk (non-emulsion) PCR amplification of products. Since such splicing occurs during the bulk phase when all cell products (e.g., many clones/unique sequences) are mixed, random (non-native) pairing could occur (see Eur J Immunol. 2013; 43(9):2507-15). The workflow would benefit from a strategy that not only reduces this occurrence, but completely prevents it. Described herein is a method in which VH and VL products do not share any common sequence, and thus the workflow is not susceptible to incidental splicing by OE-PCR during bulk re-amplification steps.
To demonstrate this, linked VH-VL ts were generated from two hybridoma clones which differ from each other by size. Native versus non-native pairing could therefore be assessed by size of products. VH and VL products from hybridoma 4G2 were generated as described in Example 3. Hybridoma 9E10 (ATCC, CRL-l729) “mini” VH and VL products were similarly produced using the same RT primers and the following PCR primers: 9E10 Mini_VL_Fwd ’-GGCAGTGGGTCTGGGACAG (SEQ ID NO: 7) mouse Kappa_ReV-Phos 5’-/5Phos/ACCAGCAGAGCTCTCACCTGGTGCAGCATCAGCCC (SEQ ID NO: 4) 9E10_VH_Fwd ’ -/5Phos/GGAACTACCGAAGGCACAGGTGAGGTGCACCTGGTGGAGTCTGGGGG (SEQ ID NO: 8) 9E 10 Mini_VH_ReV 5’ —GGATAGTGGGTGTAAGTACCACGACTACCAATG (SEQ ID NO: 9) The 4G2 VH and VL products produced were of size 400 bp and 378 bp, respectively. The mini 9E10 VH and VL products produced were of size 203 bp and 168 bp, tively. The PCR products were purified using AMPure beads.
The products were then subjected to ligase cycling reactions as described in Example 3, using cycles of thermocycling. 4G2 and 9E10 VH-VL products were ligated in separate tubes, simulating tion of natively linked products in droplets (in individual compartments). The products were analyzed by denaturing PAGE. Natively linked products of 4G2 VH-VL and of 9E10 VH-VL corresponded to sizes of 778 bp and 371 bp, respectively. Twenty cycles of ligase g were used to create products ning both ligated ts as well as non-ligated VH and VL DNA. As shown in , natively linked VH-VL products were generated for both 6G2 and 9E10.
The ligation products were purified by AMPure beads and then combined and used as te for PCR re-amplification of linked products. This step simulated recovery of products after ming linking of VH-VL in drops, ed by PCR re-amplification of linked VH—VL. By having non-ligated VH and VL DNA as template in the PCR, this provided an opportunity for DNA to become linked together during bulk re-amplification by PCR. Since the DNA was not compartmentalized by individual clones, any linkng that occurred in bulk phase would lead to non- native pairing.
PCR re-amplification was performed as described below. ” primers used to amplify linked products are as follows: 9E10 Mini_VL_Fwd ’-GGCAGTGGGTCTGGGACAG (SEQ ID NO: 7) 4G2 VL_Fwd ATCAAGATGACCCAGTCTC (SEQ ID NO: 3) 9E 10 Mini_VH_Rev 5’-GGATAGTGGGTGTAAGTACCACGACTACCAATG (SEQ ID NO: 9) Mouse IgG_CHl_rev ’-CHGATGGGGSTGTYGTTKTRGC (SEQ ID NO: 1) The PCR reactions each included: Q5 Hot Start 2X Master Mix Fwd primers (10 uM) 0.9 (each) Rev primers (10 uM) 0.9 (each) Purified ligation products 4.5 (each) Thermocycling was then performed as follows: Initial Denature Anneal Extend ration Finally, ts were analyzed by agarose gel electrophoresis. Non-natively linked products could be readily identified by their intermediate size. Specifically, 4G2-VL / 9E10-mini-VH and 9E10-mini-VL / 4G2 VH linked products would correspond to sizes of 581 bp and 568 bp, respectively. As shown in , ion of native pairing was observed when mixed during bulk re-amplification (lane 5).
Example 5: Oligo Design to Facilitate Capture of PCR Product onto Beads After RT-PCR in droplets (e.g., as described above), the PCR product can be captured onto beads within the drop to retain native matching of the VH and VL products. To facilitate efficient capture of dsDNA PCR ts onto beads, a strategy was devised to te products which have defined ssDNA at their ends. The sequence of this ssDNA is complementary to sequence of an oligo conjugated to the beads. The complementarity of these sequences thus facilitates specific and efficient capture of dsDNA PCR products.
PCR products from 4G2 cDNA were produced using PCR with the following primers and Q5 DNA polymerase. The reverse primer contains a 5’-biotin moiety to facilitate specific detection of PCR product on beads. The PCR gy was as follows: (1) (VL) 4G2 VL Fwd +PEG Mus Kap Rev biotin (2) (VL) 4G2 VL Fwd —PEG Mus Kap Rev biotin (3) (VL) 4G2 VL Fwd No_5’ Mus Kap Rev biotin (3) (VH) 4G2 VH Fwd Biotin Mus IgG-HC Rev +PEG The ing primers were used: 4G2 VL Fwd +PEG TGGATCGTTACTAATATTCGC/iSp18/GGACTCAGACACTTCCGTGCGACATCAAGATGACC CAGTCTC (SEQ ID NO: 10) 4G2 VL Fwd -PEG GTTACTAATATTCGCGGACTCAGACACTTCCGTGCGACATCAAGATGACCCAGT CTC (SEQ ID NO: 11) 4G2 VL Fwd No_5’ 5’-GGACTCAGACACTTCCGTGCGACATCAAGATGACCCAGTCTC (SEQ ID NO: 12) Mus Kap Rev biotin ’-/5BiotinTEG/ACCAGCAGAGCTCTCACCTGGTGCAGCATCAGCCC (SEQ ID NO: 13) Mus IgG-HC Rev +PEG GCAATCCATCAACGTC/iSp l8/CGTGACACATGTGGTTCAAGTACGGCHGATGGGGSTGTY GTTKTRGC (SEQ ID NO: 14) 4G2 VH Fwd Biotin ’-/5BiotinTEG/GGAACTACCGAAGGCACAGGTGAGGTCCAGCTGCAACAGTC (SEQ ID NO: PCR products were analyzed by agarose gel electrophoresis, ed by AMPure beads and fied by Nanodrop.
Amine-labeled capture oligos were conjugated to carboxylated beads L, Bangs Laboratories) using standard methods. The following oligos having sequence complementary to sequence 5’ of the PEG s in the above PCR primers were used for conjugation to beads. Beads were conjugated to VL oligo only, to VH oligo only, or to both oligos.
VL Capture GCGAATATTAGTAACGATCCAAAAAAAAAAAAAAAAAAAAA/iSp 18/iiSp1 8//iSp18//3AmM O/ (SEQ ID NO: 16) ’ -GACGTTGATGGATTGCAAAAAAAAAAAAAAAAAAAA/iSp18//iSp18//iSp18//3AmMO/ (SEQ ID NO: 17) To capture PCR product, the following reactions were setup. Purified PCR products were diluted in 0.5X SSC buffer to achieve 250 ng DNA in 25 ul final volume. Oligo-conjugated beads were washed with 0.5X SSC, and 400,000 beads were then mixed with each diluted PCR product. The bead-PCR product mix was mixed by pipet to suspend the beads, and the tubes were then placed in a thermocycler and the following program was run: Step Temp Time 1 70 0C 30 sec 2 55 0C 30 sec 3 50 0C 30 sec 4 45 °C 4 min 40 OC 4 min 6 35 °C 4 min 7 4 OC hold The samples were as follows: Blank (no capture oligo) (1) (VL) 2 Blank (no capture oligo) (2) (VL) 3 Blank (no capture oligo) (3) (VL) 4 Blank (no capture oligo) (4) (VH) VL capture (1) (VL) 6 VL capture (2) (VL) 7 VL capture (3) (VL) 8 VL capture (4) (VH) 9 VH e (1) (VL) VH capture (4) (VH) 11 VH and VL capture (1) (VL) 12 VH and VL capture (4) (VH) After temperature incubation, the samples were applied to a magnet to collect beads on the side of the tube. Supernatants were removed, and the beads were washed with 0.5X SSC buffer.
Each sample was probed for captured PCR product with 50 pl of AlexaFluor 647 IgG Fraction Monoclonal Mouse Anti-Biotin antibody (Jackson) diluted 1:1000 in PBS containing 1% BSA. After incubation with mixing for 25 minutes at room temperature, the tubes were applied to a magnet, the supernatants were removed, and the beads were washed with PBS buffer. The beads were resuspended in 50 ul PBS and then analyzed by flow cytometry.
As shown in FIGS. 5A-SB, the results demonstrate ent and specific PCR product capture by this method, with a requirement for a PEG spacer in the PCR s and an riate complementary oligo conjugated to the beads (e. g., as in sample (1)).
Example 6: Generation of Natively Paired VH-VL Product in Drops by Ligase g In this example, cells expressing VH and VL sequences of antibody 4G2 were lysed and mRNAs from the lysate were used to generate natively paired VH-VL products by ligase cycling.
Cell lysis buffer was prepared as follows: 300 ul 20% Ficoll PM400 ul 20% Sarkosyl 40 ul 5 mM EDTA 100 ul 2M Tris pH 7.5 350 ul Water 200 ul 100% ep Carboxylated COMPEL magnetic beads (Bangs Labs) were conjugated with the ing amine-modified oligos: VL Capture GCGAATATTAGTAACGATCCAAAAAAAAAAAAAAAAAAAAA/iSp 18//iSp1 8//iSp18//3AmM O/ (SEQ ID NO: 16) ’ TGATGGATTGCAAAAAAAAAAAAAAAAAAAA/iSp18//iSp18;’/iSp18//3AmMO/ (SEQ ID NO: 17) Mus_IgG_CH1_mRNA capture CTGGACAGGGATCCAKAGTTCCAAAAAAAAAAAAAAAAAAAAA/iSp18//iSp18//iSp18//3A mMO/ (SEQ ID NO: 18) Mus_Kap_mRNA capture ’ -GTGCAGCATCAGCCCGAAAAAAAAAAAAAAAAAAAA/iSp18//iSp18i/iSp18/l3AmMO/ (SEQ ID NO: 19) Oligo(dT) may also be used for mRNA capture (e.g., for capture of mRNAs encoding a VH andfor VL).
Beads were suspended in lysis buffer at a density of 2x107 beads/ml, which tates encapsulation of about 2.5 beads per drop with drop sizes used. 4G2 mouse hybridoma cells were washed with PBS and resuspended in PBS containing 0.1% BSA and 24% Optiprep at a density of 2x106 cells/m1. This density facilitates encapsulation into drops at approximately one cell per two drops. For co-encapsulation of cells and beads in drops, a 2- reagent, 100 pm diameter fluorophilic microfluidic chip (Dolomite) was used with solution flow controlled by three Mitos P-Pump pressure pumps (Dolomite). The microfluidic chip contained two input channels for s solutions and two input channels for fluorocarbon oil. Cells and beads were flowed at a rate of 10 til/min each, and fluorinated oil (HFE-7500 containing 1% urfactant (RAN Biotechnologies)) was flowed at 55 til/min. These flow rates resulted in ts of approximately 500 pl in volume. ons were collected for an hour.
The emulsions (cells and no-cell control) were applied to a heat block at 45 0C for 20 min to facilitate mRNA capture onto beads. The emulsions and beads were subsequently kept and handled at 4°C to reduce dissociation of mRNA from beads. Excess oil was removed by syringe, and 5 ml of ice-cold 6X SSC buffer containing RNAse inhibitor (1:100) was added to the emulsion. PFO (1H,1H,2H,2H-Perfluorooctanol) was added (100 ML) to the emulsion to induce drop coalescence.
The sample was mixed ghly by inversion followed by fugation at 500xg for 5 min at 4°C.
The aqueous phase was mixed by pipet, collected and then transferred to a new tube. After applying the tube to the magnet, the beads (having hybridized mRNA) were washed twice with 500 ul of ice- cold buffer (100 mM Tris HCl pH 8, 500 mM KCl, 15 mM MgC12) containing RNase inhibitor (1 : 100). The beads were then ded in 500 u] of ice-cold buffer (100 mM Tris HCl pH 8, 500 mM KCl, 15 mM MgC12) containing RNase inhibitor (1:100).
A second emulsion was prepared with the mRNA beads and RT-PCR mix. The mRNA beads were suspended in the RT-PCR bead buffer (100 ul 2X RT—PCR buffer (OneTaq ep RT-PCR, NEB), 70 ul of ep, 4 ul of RNase inhibitor, 6.4 ul of premixed 25 uM s, and 32 ul of water). Primers used were: 4G2 VL Fwd +PEG TGGATCGTTACTAATATTCGC/iSp18/GGACTCAGACACTTCCGTGCGACATCAAGATGACC CAGTCTC (SEQ ID NO: 10) Mus IgG-HC Rev +PEG GCAATCCATCAACGTC/iSp18/CGTGACACATGTGGTTCAAGTACGGCHGATGGGGSTGTY GTTKTRGC (SEQ ID NO: 14) mouse Kappa_Rev-Phos ’—/5Phos/ACCAGCAGAGCTCTCACCTGGTGCAGCATCAGCCC (SEQ ID NO: 4) 4G2 VH Fwd-Phos ’-/5Phos/GGAACTACCGAAGGCACAGGTGAGGTCCAGCTGCAACAGTC (SEQ ID NO: 5) Separately, enzyme mix was prepared (400 ul 2X RT-PCR buffer, 43 ul of enzyme diluted to 1.33X in RT-PCR buffer, 16 ul RNase inhibitor, and 341 ul water). ons for RT-PCR were generated by co-flowing beads and enzyme mix at 2.5 til/min and 7.5 ul/min, respectively, and oil (HFE7500 containing 2% fluorosurfactant) at 35 ul/min. A two- reagent 30 um diameter fluorophilic microfluidic chip (Dolomite) was used with pressure pumps to generate droplets. Under these conditions, droplets of approximately 15-35 pl in volume were , with bead occupancy of less than one bead per 5 drops. Drops were generated until all beads were encapsulated (about 30-60 minutes). The emulsions were then aliquoted into PCR tubes for thermocyling in a thermocycler with the following program: Denature 1 Cycle 55°C 50°C 45°C 40°C 12°C 30 4 min Forever sec sec The thermocycled emulsions were combined into one tube. Excess oil was removed from the bottom using a syringe. To break the drops, 100 ul of PFO was added followed by mixing and centrifugation at 2000xg for two minutes. The aqueous phase was then mixed by pipet (to suspend beads) and transferred to a new tube. The beads contained captured PCR product. The tube was d to a magnet and supernatant removed. The beads were gently washed with 500 pl of ice-cold 0.5X SSC buffer. The beads were again applied to a magnet, supernatant d and then resuspnded in 100 pl ice-cold 0.5X SSC buffer.
Samples were then prepared for ligase cycling emulsion to covalently link VH and VL products. The beads were ded in bead on (20 ul 10X Taq Ligase buffer (NEB), 117 pl 60% sucrose, and 63 ul water). Taq ligase mixture was prepared (80 ul 10X Taq Ligase buffer, 42 ul Taq Ligase, 13 ul of splint oligo (at 0.1 uM), and 665 pl water). The splint oligo used was ’ TGCCTTCGGTAGTTCCACCAGCAGAGCTCTCACCTG/3AmMO/ (SEQ ID NO: 6).
Emulsions were generated by flowing the bead solution at 2 ul/min, enzyme mix at 6 ul/min, and oil (HFE7500 containing 2% urfactant) at 35 ul/min. A two-reagent 30 um diameter fluorophilic microfluidic chip (Dolomite) was used with pressure pumps to generate droplets. Under these conditions, droplets of approximately 15-35 pl in volume were formed, with bead occupancy of less than one bead per 5 drops. Drops were generated until all beads were encapsulated (about 30-60 minutes). The emulsion was then aliquoted into PCR tubes for thermocyling in a thermocycler using the following program: Initial Denature Anneal Denaturation 1 cycle 40 cycles 2 minutes 20 30 1 Forever seconds seconds minute The thermocycled emulsions were combined into one tube. Excess oil was removed from the bottom using a e. To break drops, 100 pl of PFO was added ed by mixing and centrifugation at 2000xg for two minutes. The aqueous phase was then mixed by pipet (to suspend beads) and transferred to PCR tubes (50 ul per tube). The samples were then applied to a thermocyler preheated to 80°C to facilitate dissociation of d PCR ts hybridized to beads. After allowing beads to , the supernatant was removed and transferred to a new tube. The products were then purified using AMPure beads.
The ligated products were then reamplified in tubes by PCR using the following primers: 4G2-VL—Fwd-Reamp 5’—TGACCCAGTCTCCATCTTCA (SEQ ID NO: 23) 4G2-VH-Rev-Reamp 5’—TGTTGTTTTGGCTGAGGAGA (SEQ ID NO: 24) Component Volume (111) Thermocycling was performed as follows: Initial Denature Anneal Extend Denaturation The reamplification products were analyzed by agarose gel electrophoresis. As shown in the 4G2 cell sample yielded the intended product of linked VH-VL DNA, whereas negative control samples did not yield any product.
It is contemplated that the final PCR t, containing, e.g., natively paired VL and VH with a flexible linker sequence between in an intact open reading frame, can be cloned into a yeast surface expression vector and transformed into yeast using standard methods, resulting in a natively paired yeast y library. For example, an additional 50 bp on each terminus can be incorporated by PCR. These 50 bp match appropriate sequences in a yeast expression vector to facilitate cloning by homologous recombination in yeast, following yeast co-transformation of insert PCR product (e.g., containing ly linked VL-VH) and a linearized yeast expression vector. e 7: Self-Annealing Primers to Prevent PCR Product Capture ition from Primers PCR product capture onto beads was facilitated by tion of sticky ends (ssDNA) on a terminus of PCR products through incorporation of a spacer, such as PEG. These ssDNA portions had ce complementarity to oligos conjugated to beads, thereby tating specific hybridization. The sequences in the PCR products were incorporated by their inclusion in amplification primers. Since the PCR primers had the same sequences as the PCR product ssDNA termini, they too can be captured onto beads through specific hybridization. Such primer capture would necessarily e with PCR product capture.
Amplification of antibody repertoires by PCR typically requires a multitude of PCR s to amplify the varied antibody gene sequences. For example, primer sets recognizing V and J regions of human and mouse repertoires are often ed of about 15-20 s directed to the V regions of the heavy and light chains. The requirement for a multitude of primers leads to circumstances that exacerbate the competing effect of s on capturing PCR t onto beads. For multiplex PCR scenarios, in a given drop (compartment), a single primer (or limited number) can have complementarity to the target dy sequence within in the drop. The remaining repertoire primers will not match the target sequence. These non-matching primers would not be incorporated into amplicons during PCR, and as such, are poised to effectively compete for PCR product capture. To s this limitation, a design was ted that mitigates the ability of the unused PCR primers within a drop to compete with PCR product for capture. PCR product capture can be able, for example, under conditions of 15:1 molar ratio of unused primer to primer that generates amplicon.
Carboxylated magnetic beads were conjugated with the following amine-modified oligos, as described above.
VL_Capture GCGAATATTAGTAACGATCCAAAAAAAAAAAAAAAAAAAAA/iSp l8//iSp l 8//iSp l8//3AmM O/ (SEQ ID NO: 16) VH_Capture GACGTTGATGGATTGCAAAAAAAAAAAAAAAAAAAA/iSp lSli’iSp l8//iSp18//3AmMO/ (SEQ ID NO: 17) Mus_IgG_CH1_mRNA_capt AGGGATCCAKAGTTCCAAAAAAAAAAAAAAAAAAAAA/iSp18//iSpl8//iSpl8//3A mMO/ (SEQ ID NO: 18) p_mRNA_capt GTGCAGCATCAGCCCGAAAAAAAAAAAAAAAAAAAA/iSp18/!iSp18//iSpl8//3AmMO/ (SEQ ID NO: 19) RT-PCR reactions were set up to amplify VL sequence corresponding to 4G2 hybridoma using the following oligos. These oligos represent a design without a self-annealing ce (Non- SA) and a design with a self-annealing sequence (SA). Additionally, an oligo that has a self-annealing ’ -end but has sequence on its 5 a 3’ -end sequence that does not match the 4G2 VL or VH mRNA sequences (Mismatch) was included as an exemplary PCR primer that would not be consumed during PCR, akin to multiplex repertoire primers. The reverse primer was biotinylated to facilitate detection of captured PCR product on the bead. 4G2_VL_Fwd_Non-SA TGGATCGTTACTAATATTCGC/iSp18fGGACTCAGACACTTCCGTGCGACATCAAGATGACC CAGTCTC (SEQ ID NO: 10) Mismatch_VL_Fwd_Non-SA WO 19402 TGGATCGTTACTAATATTCGC/iSp 18/GGACTCAGACACTTCCGTGCATTGTGCTGACGCAA ACTGTTA (SEQ ID NO: 20) _Fwd_SA TGGATCGTTACTAATATTCGC/iSp 18/GAATATTAGTAACGATCCAGGACTCGGACCGACAT CAAGATGACCCAGTCTC (SEQ ID NO: 21) Mismatch_VL_Fwd_SA TGGATCGTTACTAATATTCGC/iSp 18/GAATATTAGTAACGATCCAGGACTCGGACCATTGT GCTGACGCAAACTGTTA (SEQ ID NO: 22) Mus_Kappa_ReV_Biotin iotinTEG/ACCAGCAGAGCTCTCACCTGGTGCAGCATCAGCCC (SEQ ID NO: 13) The underline designates the nnealing (complementary) sequences.
Component Volume (111) for 25 ul reaction 4G2 VL amplification primers (10 uM each) 0.5 4G2 RNA 0.1 (20 ng) 2X OneTaq One-Step RT-PCR Buffer (NEB) 12.5 OneTaq One-Step RT-PCR enzyme (NEB) 1 Oligo-conjugated beads 2 Mismatch_VL_Fwd oligo (varying concentrations, as listed below) 2.5 Water 4 Optiprep 2 RNase inhibitor 0.5 4G2 VL Amplification Fwd Primer Rev Primer Primer Mix # 1 4G2_VL_Fwd_Non-SA Mus_Kappa_ReV_Biotin # 2 4G2_VL_Fwd_SA Mus_Kappa_ReV_Biotin For competing mismatch oligo concentration used with the forward self-annealing primer (4G2_VL_Fwd_SA), the following conditions were used: Molar Ratio of Fwd Oligos Mismatch_VL_Fwd_SA (Mismatch_VL_Fwd_SA : concentration (11M) 4G2_VL_Fwd_SA) :1 30 :1 10 :1 5 1.25:1 2.5 0.625: 1 1.25 0:1 0 For competing mismatch oligo concentration used with the Fwd non-self—annealing primer (4G2_VL_Fwd_Non-SA), the following ions were used: Molar Ratio of Fwd Oligos Mismatch_VL_Fwd_Non- (Mismatch_VL_Fwd_Non-SA : SA concentration (11M) 4G2_VL_Fwd_Non-SA ) :1 10 1:1 2 0.1:1 0.2 0.01:1 0.02 0:1 0 The samples were thermocycled with the following program: After thermocycling, the samples were gently mixed by pipetting to d the beads. The samples were then placed in a thermocycler with the following program to facilitate PCR product capture onto beads: Step Temp Time 1 70 0C 30 sec 2 55 OC 4 min 3 50 OC 4 min 4 45 °C 4 min 40 OC 3 min 7 4 OC hold After the capturing ure, the tubes were applied to magnets and the atants were recovered. The supernatants were analyzed by agarose gel electrophoresis to confirm successful generation of PCR product. The beads, having captured PCR product, were washed three times with 100 pl ice-cold PBSA (1X PBS containing 1% BSA) using a magnet to sequester beads. Each sample was then incubated with 100 pl of 1:500 AlexaFluor 647 IgG Fraction Monoclonal Mouse Anti-Biotin (Jackson) in PBSA with rotation at 4°C and away from light for 45 minutes. After incubation, the beads were washed three times with 100 pl ice-cold PBSA. After a final wash, beads were resuspended with 100 pl ice-cold PBSA and analyzed with a flow cytometer for AlexaGluo647 fluorescence signal.
Mean fluorescence intensity (MFI) was determined for each sample. The sample data were plotted as a percentage of MFI relative to MFI corresponding to absence of ch oligo. As shown in FIGS. 7A-7B, only the self-annealing primers were capable of preventing PCR product e competition at high ratios of ched primer oligo to matched primer oligo. These s show that, specifically with the self-annealing primer design, the PCR product can be efficiently ed under conditions of 15-fold more unused primer — conditions that, for example, simulate multiple antibody repertoire conditions.
ORATION BY REFERENCE All publications, patents, and Accession numbers ned herein are hereby incorporated by reference in their entirety as if each individual publication or patent was specifically and individually indicated to be incorporated by reference.
EQUIVALENTS While specific embodiments of the subject invention have been discussed, the above specification is illustrative and not restrictive. Many variations of the invention will become apparent to those skilled in the art upon review of this specification and the claims below. The full scope of the invention should be ined by reference to the claims, along with their full scope of equivalents, and the specification, along with such variations.

Claims (47)

What is claimed is:
1. A method of making a nucleic acid sequence comprising a sequence that encodes a heavy chain element (HC element) of an antibody heavy chain variable region (HCVR) and a light chain element (LC t) of an antibody light chain variable region , and wherein the HCVR and LCVR are matched, the method comprising: a) acquiring an isolated production reaction site, comprising: i) a heavy chain (HC) strand, wherein the HC strand is a strand of a heavy chain double-stranded cDNA (HC ds cDNA) comprising a segment that encodes the HC element of 10 the HCVR from a cell; and ii) a light chain (LC) strand, wherein the LC strand is a strand of a light chain double- stranded cDNA (LC ds cDNA) comprising a segment that encodes the LC t of the LCVR from the cell, and b) covalent linking of the HC strand to the LC strand, 15 n the isolated production reaction site does not comprise a nucleic acid encoding an HCVR or an LCVR from a cell other than the cell, thereby making the nucleic acid sequence.
2. The method of claim 1, wherein the HC element comprises, or consists of, a heavy chain 20 variable region sequence (HCVRS), or an antigen binding nt thereof.
3. The method of claim 1 or 2, wherein the LC element comprises, or consists of, a light chain variable region sequence ), or an antigen binding fragment thereof. 25
4. The method of any of claims 1-3, wherein the nucleic acid sequence is configured such that, when expressed, the HC element and the LC element form a functional antigen binding molecule in vilro, ex vivo, or in vivo.
5. The method of any of claims 1-4, wherein acquiring the isolated production reaction site 30 ses: a) acquiring a capture substrate bound to: (i) a first -stranded cDNA (ds cDNA) comprising a strand that is complementary to a first mRNA that encodes the HCVR from the cell; and (ii) a second ds cDNA sing a strand complementary to a second mRNA 35 encoding the LCVR from the cell, to produce a loaded e substrate, and b) maintaining the isolated production reaction site under ions that allow amplification of the first and second ds cDNAs, to produce: a plurality of HC ds cDNAs comprising the segment that encodes the HC element of the HCVR from the cell; and a plurality of LC ds cDNAs comprising the segment that encodes an LC element of the LCVR from the cell.
6. The method of any of claims 1-5, wherein the e ate comprises a bead and a moiety which binds to cDNA.
7. The method of any of claims 1-6, wherein the ed production reaction site comprises a 10 reagent mixture suitable for producing, from the first and second mRNAs, a first ds cDNA comprising the segment that encodes the HC element of the HCVR of the cell, and a second ds cDNA comprising a t that encodes the LC element of the LCVR of the cell.
8. The method of any of claims 5-7, wherein the first and second ds cDNAs are ed in 15 the presence of primers, wherein at least one of the primers comprises a first member, a second member, and a nucleotide modification between the first and second members, wherein the nucleotide modification reduces DNA synthesis.
9. The method of claim 8, wherein the nucleotide modification comprises an insertion of a 20 spacer between two adjacent nucleotides or a modification to a ribose.
10. The method of claim 8 or 9, wherein the first member is capable of annealing with the second member in the same primer or a different , forming a double-stranded structure comprising a duplex region of 4, 5,6, 7, 8, 9, 10, ll, 12, 13, 14, 15, 16, 17, 18, 19, 20, more 25 basepairs.
11. The method of any of claims 8-10, n at least one of the primers is phosphorylated and comprises a sequence encoding at least a portion of a linker sequence, or a complementary sequence thereof.
12. The method of any of claims 1-1 1, n the HC ds cDNA comprises a 5’ overhang and a blunt end and the LC ds cDNA comprises a 5’ overhang and a blunt end.
13. The method of any of claims 1-12, wherein the HC strand and the LC strand are 35 covalently linked to produce a single stranded nucleic acid sequence, wherein the HC and LC strands are both sense strands or both antisense strands.
14. The method of any of claims 1-13, wherein the covalent linking occurs in an isolated linkage reaction site comprising a ligase.
15. The method of any of claims 1-14, wherein the HC strand and the LC strand are covalently linked in the presence of a splint oligonucleotide, wherein the splint oligonucleotide is hybridized to a ce comprising the junction of the HC strand and the LC strand to form a duplexed region at the site of linkage.
16. The method of claim 15, wherein the splint oligonucleotide comprises a modification that 10 inhibits DNA synthesis.
17. The method of any of claims l-l6, further sing, prior to acquiring the isolated production reaction site, ing an mRNA loaded capture substrate comprising: a) acquiring an isolated cell reaction site, comprising: 15 i) a cell; and ii) a capture ate capable of binding a first mRNA encoding an HCVR from the cell and a second mRNA ng an LCVR from the cell; and b) maintaining the ed cell reaction site under conditions that allow lysis of the cell and binding of the capture substrate with the first mRNA and the second mRNA to form the mRNA 20 loaded capture substrate, wherein the isolated cell reaction site does not include a nucleic acid encoding an HCVR or an LCVR from a cell other than the cell.
18. The method of claim 17, further sing releasing the mRNA loaded capture substrate 25 from the isolated cell reaction site in the presence of a poly(dA) or poly(dT) oligonucleotide.
19. The method of any of claims 1-18, further comprising amplifying the nucleic acid sequence. 30
20. The method of any of claims 1-19, further comprising cing all or a portion of the nucleic acid sequence.
21. The method of any of claims 1-20, r comprising inserting all or a portion of nucleic acid sequence into a vector.
22. The method of any of claims 1-21, comprising expressing the nucleic acid sequence to produce a polypeptide comprising the segment that encodes the HC element of the HCVR, and the segment that encodes the LC element of the LCVR.
23. The method of claim 22, further comprising contacting the ptide with an antigen and determining if the polypeptide binds the antigen.
24. A method of making a library comprising a plurality of unique members, the method comprising: 10 making the plurality of members by the method of any of claims 1-23, n each of the members comprises a sequence that encodes a heavy chain element (HC t) of a heavy chain variable region (HCVR) and a light chain element (LC element) of a light chain variable region (LCVR), wherein the HCVR and the LCVR are matched, and wherein each unique nucleic acid sequence of the ity comprises an HC element and an LC element from a 15 ent unique cell, thereby making the library.
25. The method of claim 24, wherein the library comprises one, two, three, or all of the following properties: 20 a) the plurality of unique s ses at least 104, 105, 106, 107, 108, or 109 unique members; b) the plurality of unique members comprises 104 to 109, 104 to 108, 104 to 107, 104 to 106, 104 to 105, 108 to 109, 107 to 109, 106 to 109, 105 to 109, 105 to 103, 106 to 107, 104 to 105, 105 to 106, 106 to 107, 107 to 108, or 108 to 109 unique members; 25 c) at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%, of the members in the library are unique members,; or d) less than 20%, 15%, 10%, 5%, 4%, 3%, 2%, or 1%, of the members in the library are unique members. 30
26. A library made by the method of claim 25.
27. The library of claim 26, wherein the library is a display library.
28. A method of making a binding polypeptide, the method comprising: 35 a) acquiring a library of claim 26 or 27; and b) sing a polypeptide encoded by a unique nucleic acid of the library.
29. The method of claim 28, further comprising contacting the polypeptide with an antigen and obtaining a nucleic acid that s a polypeptide that binds the n.
30. A method of making a nucleic acid sequence comprising a sequence that encodes an or chain element (AC element) of a T-cell receptor (TCR) or chain le region (ACVR) and a [5 chain element (BC element) of a TCR [3 chain variable region (BCVR), and wherein the ACVR and BCVR are matched, the method comprising: a) acquiring an isolated tion reaction site sing: i) an or chain (AC) , wherein the AC strand is a strand of an a chain double- 10 stranded cDNA (AC ds cDNA) comprising a segment that s an AC element of the ACVR from a cell; and ii) a [3 chain (BC) strand, wherein the BC strand is a strand of a [3 chain double- stranded cDNA (BC ds cDNA) comprising a segment that encodes a BC element of the BCVR from the cell, and 15 b) covalent linking of an AC strand to a BC strand, wherein the isolated production reaction site does not comprise a nucleic acid encoding a BCVR or an ACVR from a cell other than the cell, thereby making the nucleic acid sequence. 20
31. The method of claim 30, wherein the nucleic acid sequence is configured such that, when expressed, the AC element and the BC t form a functional antigen binding molecule.
32. The method of claim 31, wherein the covalent linking occurs in an isolated linkage on site comprising a ligase.
33. A method of making a library comprising a plurality of unique members, the method comprising: making the plurality of members by the method of any of claims 30-32, n each of the members comprises a sequence that encodes a or chain element (AC 30 element) of a or chain variable region (ACVR) and a [3 chain element (BC element) of a [3 chain variable region (BCVR), wherein the ACVR and BCVR are matched, and wherein each unique nucleic acid sequence of the plurality comprises an AC element and a BC element from a different unique cell, thereby making the library.
34. A library made by the method of claim 33.
35. A method of making a binding polypeptide, the method comprising: a) acquiring the library of claim 34; and b) expressing a polypeptide encoded by a unique nucleic acid of the library.
36. A method of making a nucleic acid sequence comprising a sequence that s an 7 chain element (GC element) of a TCR 7 chain variable region (GCVR) and a 5 chain element (DC element) of a TCR 5 chain variable region (DCVR), and wherein the GCVR and DCVR are d, the method comprising: a) acquiring an isolated production reaction site, sing: 10 i) a y chain (GC) , wherein the GC strand is a strand of a 7 chain double- stranded cDNA (GC ds cDNA) comprising a segment that encodes a GC element of the GCVR from a cell; and ii) a 5 chain (DC) strand, wherein the DC strand is a strand of a 5 chain double- ed cDNA (DC ds cDNA) comprising a segment that encodes a DC element of the 15 DCVR from the cell, and b) nt linking of a GC strand to a DC strand, wherein the isolated production reaction site does not comprises a nucleic acid encoding a DCVR or a GCVR from a cell other than the cell, thereby making the nucleic acid sequence.
37. The method of claim 36, wherein the nucleic acid sequence is ured such that, when expressed, the GC element and the DC element form a functional antigen binding le.
38. The method of claim 36 or 37, wherein the covalent linking occurs in an isolated linkage 25 reaction site comprising a ligase.
39. A method of making a library comprising a ity of unique s, the method comprising: making the plurality of members by the method of any of claims 36-38, 30 wherein each of the members ses a sequence that encodes a 7 chain element (GC element) of a 1/ chain variable region (GCVR) and a 5 chain element (DC element) of a 5 chain variable region (DCVR), wherein the GCVR and DCVR are matched, and wherein each unique nucleic acid sequence of the plurality comprises a GC element and a DC element from a different unique cell, 35 thereby making the library.
40. A library made by the method of claim 39.
41. A method of making a binding polypeptide, the method comprising: a) acquiring the library of claim 40; and b) expressing a polypeptide encoded by a unique nucleic acid of the library.
42. An isolated tion reaction site, comprising: a) a heavy chain (HC) strand, wherein the HC strand is a strand of a heavy chain double- ed cDNA (HC ds cDNA) comprising a segment that encodes an HC element of the HCVR from a cell; and 10 b) a light chain (LC) strand, wherein the LC strand is a strand of a light chain double-stranded cDNA (LC ds cDNA) comprising a segment that encodes an LC element of the LCVR from the cell, c) a primer comprising a first member, a second member, and a nucleotide modification between the first and second members, wherein the nucleotide modification s DNA synthesis, wherein the HCVR and LCVR are d, and wherein the isolated production reaction site 15 does not comprise a c acid encoding an LCVR or an HCVR from a cell other than the cell.
43. The isolated tion reaction site of claim 42, wherein the first member is capable of annealing with the second member in the same primer or a different , forming double-stranded structure comprising a duplex region of 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, more basepairs. .
44. A self-annealing oligonucleotide comprising a first member, a second , and a spacer located between the first and second members, wherein the first member is capable of annealing with the second member in the same oligonucleotide or a different oligonucleotide, forming 25 a hairpin structure or a double-stranded structure comprising a duplex region of 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19,20, more basepairs.
45. The oligonucleotide of claim 44, wherein the spacer is a flexible spacer or a PEG spacer. 30
46. An isolated linkage reaction site, comprising: a) a heavy chain (HC) strand, wherein the HC strand is a strand of a heavy chain double- stranded cDNA (HC ds cDNA) comprising a segment that s an HC element of the HCVR from a cell; b) a light chain (LC) strand, wherein the LC strand is a strand of a light chain -stranded 35 cDNA (LC ds cDNA) comprising a segment that encodes an LC element of the LCVR from the cell; c) a splint oligonucleotide that is capable of hybridizing to a sequence comprising the junction of the HC strand and the LC strand, to form a duplexed region at the site of linkage, wherein the HCVR and LCVR are matched, wherein the HC strand and the LC strand are covalently , and wherein the isolated linkage reaction site does not comprise a nucleic acid encoding an HCVR or an LCVR from a cell other than the cell,
47. The isolated linkage reaction site of claim 46, further comprising a ligase. Wage ammo, Wommc mfi Exam g Ex? mfi ”Exam E $0 E 30. E $0 3&5 E 85¢.me 3&5 SE35 ,8st .Nm mm 850.35 3% E Em 3 3% gm $333 £03 3cm Eon” mg E 0w («Emu nmfiommfi Eu 3 flung {2% cc 3 @338 £00m flag.9 so comma: Emu muSSQ Sufiwoa Emma: (2% 4&2me flu mu 3cm Wmfimmfi Embmgsm mv mam mm SE33 MI mnSQm u: 3333 wnfioa Wm Egommfi 3335 m: {Emu B E fiéu me + u: <2Qu Wmfimmfi 3 we «2% “E mumbfifi casmmfi: flu mam um E 95» mafia E <23.» hmfimmow .3 dzmu mo 39$me guise @355 QE< QE< m: mu. mu mm mm“ mm”. mm ,3 Emfi Na. mwgmzm 3 £338 83mg QE<.mm @335H Emfi c3 mvcgm 3 36% 8338 mm- 3:553 Emfi mm mvgmbm 3 338 $358 . .. WE .E 3 Eek 8 «ZmE Ema. Ow {ZmE Eat 3 E Fm E mmmmmm; gmbmgsm we wumflse mmammmh .mwmmeSm “5 mm «Egg cemmmofl .wme «Emu {Emu E EN Emam 33mm.“ .mwmsmomnm Emu cm 3mm <ZmE @338 munwei £03.39 SEE <2me .U cmu wkdwfimu $33th £55:QO ggfimmm mq. 33%th 838a .goWEWQm gowumwm 38% ““0Wmhfinwu $3 £338 am a Em <sz @3353 commmomm @333 .E gm Smbmga SUBSTITUTE SHEET (RULE 26) * 3 0339305 ccnjugated *1 03390 for mRNA capture {paw-{cm} .. HQ mRNA /* LC mRNA Beads; with ed mRNA in aqueaus phaga HQ 2% SUBSTITUTE SHEET (RULE 26) Spacer LCVR sequanae Compiementaryta Universaipriming bead ca ptur‘e oiiga sequence Lin ker Sequencas LCVR pmduct HC‘JR produqt SUBSTITUTE SHEET (RULE 26) SUBSTITUTE SHEET (RULE 26) €35 Fiexibfe LCVP‘ HCVR __Linker ........... Séq fer Seq far homeiagous hamoiogms remmbina’cion ination Fifi-3., 2E3 SUBSTITUTE SHEET (RULE 26) WO 19402 SUBSTITUTE SHEET (RULE 26) —amp§e-L 462 VH—VL m 800 m 600 9E1 0 Mini VH-VL —-— 460 462 VH—VL : W8 hp Mixed VH—VL product :2 5681581 bp 9E1 0 Mini VH—VL : 371 hp HG. 4B SUBSTITUTE SHEET (RULE 26) Primer PCR Produd VL sequence} {3} EZZF-- fi} (43 VH sticky-end PCR product Ffiifisfgik 16000%qnmmwmmmmnmmmmnm ‘" 8900€”_nwmmmmmnmnmwmmnn 4000?“ .....M "w M _____ EPCRFVGdUct IflflllfifllflfifilIflfllIflfilflfiflllflflfllflfllllflliflfilIflll SEEM-Gigs Biank VL-Qiigo VH-oiige ‘v’H-i-VL-aiégo ....................................................................................................................................................................................................................... SUBSTITUTE SHEET (RULE 26) 462 PCR NTC SUBSTITUTE SHEET (RULE 26) 10l10 PCR Praduct Capture 4mm Beads Uging Non‘Seifu Capture Anneaimg PCB 01290 123 ....................................................................... g ma~~~ Product g; saw g 68-- :1 45:2»- PCR a 28-~ 1% (3 VL 0:1 (101:1 Cam 1:1 5:”: Ratio of ah Oiiga : Matched {)Eigfl HQ. 72% PER Product Capture (into Beads Using Ssh“— Capture Anneaiing PCR Géigo 193W ...................... Pmduct PCR ‘Nm‘maiized 8Q .......... ............... £3 C3 49” .... 29m ............. VL {3 9:? 0952521 1.25:? 25:1 5:? ”35:1 Ratio 0? Mismamh Giiga : Matched Oiigo HQ. 7% SUBSTITUTE SHEET (RULE 26)
NZ795976A 2016-12-23 2017-12-22 Binding polypeptides and methods of making the same NZ795976A (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US62/438,712 2016-12-23

Publications (1)

Publication Number Publication Date
NZ795976A true NZ795976A (en) 2022-12-23

Family

ID=

Similar Documents

Publication Publication Date Title
TWI829621B (en) Anti-human vista antibodies and use thereof
JP6731403B2 (en) Compositions and methods for use in enhancing immune response and cancer treatment
AU2015357463B2 (en) Identification of VSIG8 as the putative vista receptor and its use thereof to produce vista/VSIG8 modulators
US20240141334A1 (en) Binding polypeptides and methods of making the same
KR20190057366A (en) Chimeric antigen receptor for the treatment of cancer
CN117285641A (en) Guidance and navigation control proteins and methods of making and using the same
KR20170134642A (en) CD20 therapy, CD22 therapy, and combination therapy with CD19 chimeric antigen receptor (CAR) -expressing cells
CN110891650A (en) Guidance and navigation control proteins and methods of making and using same
AU2013278843A1 (en) LSR antibodies, and uses thereof for treatment of cancer
KR20200022447A (en) Dosage regimens of anti-TIM-3 antibodies and uses thereof
WO2019137397A1 (en) Pd-l1 antibody, antigen-binding fragment thereof, and pharmaceutical use thereof
JP2020530306A (en) Multispecific antibody and its preparation and usage
JP2020530777A (en) Multispecific antibody and its preparation and usage
WO2018156735A1 (en) Bispecific antibody for cancer immunotherapy
NZ795976A (en) Binding polypeptides and methods of making the same
NZ753978A (en) Binding polypeptides and methods of making the same
US20220185911A1 (en) Therapeutic antibodies for treating lung cancer
RU2809160C2 (en) Types of combination therapy using chimeric antigen receptors and pd-1 inhibitors
TW201625697A (en) Compositions and methods of use for augmented immune response, and cancer therapy
TW202328196A (en) Treatment and prevention of cancer using vista antigen-binding molecules
IL256261A (en) Hide1 compositions and methods