Movatterモバイル変換


[0]ホーム

URL:


WO2024249362A1 - Rare antibody phage isolation and discrimination (rapid) biological panning - Google Patents

Rare antibody phage isolation and discrimination (rapid) biological panning
Download PDF

Info

Publication number
WO2024249362A1
WO2024249362A1PCT/US2024/031136US2024031136WWO2024249362A1WO 2024249362 A1WO2024249362 A1WO 2024249362A1US 2024031136 WUS2024031136 WUS 2024031136WWO 2024249362 A1WO2024249362 A1WO 2024249362A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
interest
antibodies
target
phages
Prior art date
Application number
PCT/US2024/031136
Other languages
French (fr)
Inventor
Dong Hee Chung
Charles S. Craik
Nicholas J. YOUNG
Original Assignee
The Regents Of The University Of California
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 The Regents Of The University Of CaliforniafiledCriticalThe Regents Of The University Of California
Publication of WO2024249362A1publicationCriticalpatent/WO2024249362A1/en

Links

Classifications

Definitions

Landscapes

Abstract

The present disclosure provides methods and systems for identifying rare high-affinity antibodies against challenging targets. In certain aspects, these methods and systems include novel profiling techniques for monitoring the enrichment of a biopanning campaign. In certain aspects, these methods and systems include novel screening techniques for identifying, ranking, and prioritizing candidate binding antibodies. In certain aspects, these methods and systems are coupled together into a single novel identification pipeline, wherein the disclosed profiling techniques are used to selectively isolate the most promising population of binding antibodies and the disclosed screening techniques subsequently perform screening in a discriminatory fashion. In certain aspects, these methods and systems allow for the identification of antibodies against antigens previously beyond the scope of immunization.

Description

RARE ANTIBODY PHAGE ISOLATION AND DISCRIMINATION (RAPID) BIOLOGICAL PANNING
STATEMENT AS TO FEDERALLY SPONSORED RESEARCH
This invention was made with government support under Grant No. U54AI170792 awarded by the National Institutes of Health, and Grant No. DBI- 1548297 awarded by the National Science Foundation. The government has certain rights in the invention.
CROSS-REFERENCE TO RELATED APPLICATIONS
This application claims the benefit of priority to U.S. Provisional Application Serial Number 63/505,665, filed June 1, 2023, and U.S. Provisional Application 63/584,750, filed September 22, 2023, the contents which are hereby incorporated by reference in their entirety.
INCORPORATION BY REFERENCE OF SEQUENCE LISTING PROVIDED AS A SEQUENCE LISTING XML FILE
A Sequence Listing is provided herewith as a Sequence Listing XML file, “UCSF- 717WO_SEQ_LIST.xml”, created on May 24, 2024, and having a size of 4,597 bytes. The contents of the Sequence Listing XML file are incorporated by reference herein in their entirety.
INTRODUCTION
Antibodies (Abs) have and continue to be invaluable tools for biological research. In particular, the utility of rapidly generated, high-quality Abs has been highlighted in the 2020 SARS-CoV-2 pandemic as attractive therapeutic interventions. While immunization has been the traditional method for Ab discovery, biopanning using synthetically displayed Ab libraries (i.e., phage display and yeast surface display) have expanded the field and allowed for non-protein antigens (Ags) such as DNA and RNA to be targeted.
Standard phage display biopanning methods largely consist of two stages: (1) Iterative rounds of washing and amplification of displayed Abs bound to immobilized Ag to achieve an enriched population of higher- affinity binders; (2) Random screening of enriched pools of displayed Abs for the identification of high- affinity Ab clones (hits). Notably, the in vitro selection and screening nature of the method enables the identification of high-affinity Abs in conditions otherwise impossible with immunization (i.e., changes in pH, temperature, oligomeric complex states, ligand states etc.),
Despite these attractive features of biopanning, it remains challenging to identify, and characterize selective, high-affinity Abs where their prevalence is extremely low. Examples of these Ags include those that exhibit low antigenicity, or conformational heterogeneity (e.g., Ags that are conformationally dynamic). Poor enrichment towards Ag-Ab binding allows additional factors such as amplification discrepancies of phagemid-containing E. coli, and inconsistent display propensities of phage, to undermine the enrichment process resulting in an overall depletion of promising binders with continued rounds of selection. In turn, a larger number of clones need to be screened and prioritized, and as this process largely relies on stochastic clone picking, the vast majority of candidate clones, and potentially rare high-affinity binders, are left unexamined.
SUMMARY
The present disclosure provides methods and systems for identifying rare high-affinity antibodies against challenging targets. In certain aspects, these methods and systems include novel profiling techniques for monitoring the enrichment of a biopanning campaign. In certain aspects, these methods and systems include novel screening techniques for identifying, ranking, and prioritizing candidate binding antibodies. In certain aspects, these methods and systems are coupled together into a single novel identification pipeline, wherein the disclosed profiling techniques are used to selectively isolate the most promising population of binding antibodies and the disclosed screening techniques subsequently perform screening in a discriminatory fashion. In certain aspects, these methods and systems allow for the identification of antibodies against antigens previously beyond the scope of immunization. In addition to the identification of rare antibodies against challenging targets, the profiling and screening techniques of the disclosed methods and systems allow for identification of antibodies with lower kinetic off-rates, enhanced screening sensitivity, and a reduction in time to identify antibodies of interest.
In one aspect, methods for identifying an antibody with affinity to a target of interest by using bio-layer interferometry (BLI) are provided. Aspects of the methods include: contacting a sensor tip including the target of interest immobilized thereon with a first aliquot of a solution including multiple copies of a first antibody; irradiating the sensor tip with an incident light and measuring a first reflected light, wherein a wavelength shift of the first reflected light compared to the incident light is indicative of binding of the first antibody to the target of interest immobilized on the sensor tip; contacting the sensor tip with a second aliquot of the solution including multiple copies of the first antibody, wherein the second aliquot further includes a second antibody that binds to the first antibody; and irradiating the sensor tip with the incident light and measuring a second reflected light, wherein a wavelength shift of the second reflected light compared to the incident light and/or the first reflected light is indicative of binding of the second antibody to the first antibody bound to the sensor tip, wherein presence of: (a) the wavelength shift of first reflected light compared to the incident light and (b) the wavelength shift of the second reflected light compared to the incident light and/or the first reflected light identifies the first antibody as having affinity to the target of interest.
In certain embodiments, the method further includes contacting a buffer with the sensor tip, wherein the buffer liquid includes a lower concentration of first antibody than the solution, e.g., does not contain detectable level of the first antibody. In some embodiments, a binding kinetic parameter of the copies to the target of interest is determined using tip measurements generated while the tip is in the buffer. In some embodiments, false positives are determined using tip measurements generated while the tip is in the second aliquot. In some embodiments, whether or not the sensor tip is saturated by antibody copies is determined using tip measurements generated while the tip is in the first aliquot of the solution.
In certain embodiments, the screening is performed for each antibody of a plurality of different antibodies. In some embodiments, each of the plurality of antibodies are ranked based on a binding kinetic parameter predicted for each of the different antibodies to the target of interest. In some embodiments, the binding kinetic parameter is a dissociation rate constant.
In certain embodiments, the plurality of antibodies is identified and isolated using the profiling methods of the present invention, e.g., as discussed in greater detail below.
In another aspect, methods of identifying antibodies with affinity to a target of interest are provided. In some embodiments, the methods include profiling a biopanning campaign in order to isolate a plurality of antibodies with affinity to a target of interest. Aspects of the methods include: conducting a first round of biopanning including isolating phages displaying antibodies that bind to the target of interest and fluorescently labeling the phages to generate a first labeled population of phages; conducting a second round of biopanning including isolating phages displaying antibodies that bind to the target of interest and fluorescently labeling the phages to generate a second labeled population of phages; separately contacting the first and the second labeled population of phages with the target of interest immobilized on beads to produce bead-phage complexes; analyzing the complexes using flow cytometry to generate a first fluorescence profile of the complexes produced from the first labeled population of phages and a second fluorescence profile of the second labeled population of phages, using the first and second profiles to identify antibodies with affinity to the target of interest.
In certain embodiments, the profile includes a histogram representing the frequency of complexes having different fluorescence intensities. In some embodiments, a median fluorescence intensity (MFI) and/or a measure of the variability of the profile such as, e.g., a standard deviation (SD) or coefficient of variation (CV) are calculated for one or more components of each profile. In some embodiments, the high affinity antibodies correspond to a component of the profile having a high MFI.
In certain embodiments, the methods further include sorting the complexes based on a level of fluorescent phages bound to the beads, thereby separating complexes exhibiting a threshold level of fluorescence from complexes exhibiting fluorescence below the threshold level.
In certain embodiments, the high affinity antibodies identified using the profiling methods of the present invention are individually screened to identify antibodies with a high affinity to the target of interest using the bio-layer interferometry (BLI) screening methods of the present invention.
In another aspect, a system for identifying antibodies with affinity to a target of interest is provided, the system including: a processor including memory operably coupled to the processor wherein the memory includes instructions stored thereon, which when executed by the processor, cause the processor to: obtain raw bio-layer interferometry data, wherein the interferometry data includes sequential wavelength shift measurements resulting from interactions between a sensor tip including a target of interest, copies of an antibody, and tag binding antibodies; section the interferometry data into two consecutive segments; fit a one phase exponential association curve to raw data from the first segment, wherein the sequentially later end of the one phase exponential association curve includes a first slope; fit a line to the second segment data, wherein the fitted line includes a second slope; and compare the first slope to the second slope, wherein the antibody copies are identified as having affinity to the target of interest if the second slope is greater than the first slope.
In certain embodiments, the memory includes instructions stored thereon, which when executed by the processor, cause the processor to refine raw data from the second segment by extrapolating the association curve fit to the first segment through the second segment and subtracting the extrapolated association curve from the raw data of the second segment, wherein the line is fit to the refined second segment data.
In certain embodiments, the memory includes instructions stored thereon, which when executed by the processor, cause the processor to section the interferometry data into three consecutive segments, wherein a one phase exponential decay curve is fit to raw data from the third segment.
In certain embodiments, the memory includes instructions stored thereon, which when executed by the processor, cause the processor to determine a goodness of fit metric for each fitted curve. In some embodiments, the goodness of fit metric is an r-squared value.
In certain embodiments, the memory includes instructions stored thereon, which when executed by the processor, cause the processor to detect negatives of the antibody copies not binding to the target of interest, false positives of non-specific binding proteins binding to the target of interest, and/or hits of the antibody copies binding to the target of interest. In some embodiments, negatives and false positives are detected by determining if the slope of the line fit to the second segment is below a predetermined threshold value and/or if the r-squared value of the line fit to the second segment is below a predetermined threshold value. In some embodiments, hits are detected by determining if the slope of the line fit to the second segment meets of exceeds a predetermined threshold value and if the r-squared value of the line fit to the second segment meets or exceeds a predetermined threshold value. In some embodiments, whether or not the sensor tip is saturated by antibody copies is determined by calculating a slope of the extrapolated association curve and determining if the slope of the extrapolated association curve meets or exceeds a predetermined threshold value.
In certain embodiments, the system is configured to identify antibodies with affinity to the target of interest for a plurality of inputs each including sequential wavelength shift measurements resulting from interactions between a sensor tip including a target of interest, copies of an antibody, and tag binding antibodies. In some embodiments, the copies for each input are of a different antibody. In some embodiments, the inputs are ranked based on a binding kinetic parameter predicted for each of the different antibodies to the target of interest. In some embodiments, the binding kinetic parameter is a dissociation rate constant.
In certain embodiments, the system further includes a bio-layer interferometry sensor tip configured to generate the raw bio-layer interferometry data and transmit it to the processor, the sensor tip including: a surface including the target of interest; a light source configured to irradiate the surface; and a detector configured to detect an interaction between antibodies and the target of interest.
In another aspect, a non-transitory computer-readable medium is provided, the non- transitory computer-readable medium including the instructions of the memory of the systems described herein.
BRIEF DESCRIPTION OF THE FIGURES
FIG. 1 Schematic of RAPID biopanning for the identification of rare high-affinity binders against challenging targets. RAPID biopanning follows a Label-Profile-Sort-Screen pipeline. First, phage libraries from rounds of biopanning are individually FITC (yellow star) labeled and each incubated with antigen-beads (beige antigen, grey bead). Subsequent analysis with flow cytometry profiles the progression of the biopanning campaign and accurately identifies the most enriched round for antigen- antibody binding. For phage displayed Fab (ph-Fab) libraries that exhibit extremely low frequencies of high-affinity antibodies (Abs) against antigens (Ags), fluorescent activated sorting is performed to isolate specific populations of ph-Fabs that contain higher frequencies of high affinity binders. A novel BLI method is subsequently employed to rapidly screen candidate clones in a discriminatory matter, thereby prioritizing promising hits for further investigations. For comparison, standard biopanning using magnetic beads (beige antigen, red bead) is depicted in the upper left comer.
FIGS. 2A to 2F flow cytometry of four ph-Fabs with ARS1620-V7 (VVVGACGVGK (SEQ ID NO:1)). (A) Pull down titers of ph-PlA4, ph-PICl, ph-PlH6, and ph-P2Fl l. FITC labeled and unlabeled ph-Fab exhibit similar phage titers. (B) FITC labeling of Ml 3, ph-Pl A4, andph-P2Fl l. (C) Flow cytometry analysis of ph-PICl, ph-PlH6, ph-P2Fl l, and ph-Pl A4 bound to Ag-bead. (D) Normalized median fluorescent intensity (MFI) of flow cytometry data from (D). Importantly, the normalized MFI trends match with the labeled titer trends in (A), ensuring ph- Fabs are labeled with no biases and interactions between Ag and displayed Fabs are not significantly affected by FITC labeling. (E) RAPID flow cytometry profiling with different percentages of ph-PlA4 and M13 (FITC labeled). (F) An increase in ph-Pl A4 concentration shows an increase in normalized MFI with an observable shift from flow cytometry starting at 10 percent ph-Pl A4.
FIG. 3 depicts RAPID fluorescent activated sorting of fluorescently labeled ph-Fab libraries. RAPID fluorescent activated sorting allows for the isolation of higher- affinity ph-Fab populations. Iterative rounds of soiling can result in the enrichment of ph-Fab libraries to be rebiased towards Ag-Fab binding where severe growth/display propensity biases occur in standard biopanning. ph-Fab libraries are first FITC labeled and incubated with Ag-Beads. Non-binding phage are removed by washing, and Ag-Bead-fluorescent ph-Fab complex is sorted and bead populations containing high fluorescence are isolated.
FIGS. 4A to 4D biolayer Interferometry Antibody Screen (BIAS). (A) BIAS scheme. Representative BLI sensograms of each possible scenario (Hit, False positive, Negative) are shown on the right, (i) Association- 1 : Biological tips bound with Ag are transferred to crude PPEs induced for expression, (ii) Association-2; tips are transferred to wells containing identical PPE sample from Association- 1 + anti-myc IgG, (iii) Dissociation: tips from (ii) are transferred to wells with no PPE and no IgG. The Association-2 step distinguishes hits versus false positives. (B)-(D) BLI sensograms of BIAS performed with ARS1620 and P1A4 spiked samples. (B) P1A4 (250 nM) spiked in PBS. A signature linear curve is observed in Assco-2 where the anti-myc antibody (9E10) is present, while this is not observed with the control with no 9E10. (C) P1A4 (250 nM) spiked in TGI -PPE. Results are similar as (C). (D) Concentration series of P1A4 spiked in TGI -PPE (10 - 300 nM).
FIGS. 5 A to 5D rare antibody phage isolation and discrimination (RAPID) biopanning of Carboxyl terminus of Hsp-70 interacting protein (CHIP). (A) PhAB flow from Rounds 1-4. Round 3 shows the highest fluorescent distribution shift. (B) Comparison of Biolayer interferometry antibody screen (BIAS) koff and biolayer interferometry (BLI) measured koff at 500 nM. (C) BIAS koff of candidate hits. (D) Candidate hit screening using dot blot and BIAS. Dot blot hits are circled in black where hits that overlap with BIAS hits are circled with a thicker boarder. BIAS Hits, False positives, and Negatives are shown according to the legend. FIGS. 6A to 6H antibody discovery campaigns of Carboxyl terminus of Hsp-70 interacting protein (CHIP) and Gaq. (A)-(F) Full BIAS curves for six candidate clones. (G)-(H) Gaq hits from BATCH. Clones with potentially inaccurate koff predictions are flagged ( ow expression #- unsatruated association- 1, 1-poor exponential fit for dissociation). Assoc- 1, Assoc-2, Dissoc steps are marked with green, blue, purple respectfully.
FIGS. 7 A to 7B antibody discovery campaign of CS3D. (A) Zoom in overlay of PhAB flow of rounds 1-5. A shoulder distribution is observed. (B) Normalized MFI of PhAB flow of rounds 1-5.
FIG. 8 increasing concentrations of biotin-FITC to streptavidin beads. A maximal distribution shift is observed starting at 1 pM biotin-FITC immobilization with no overlap to the negative control with no phage.
FIGS. 9 A to 9D depict RAPID Flow cytometry enrichment profiling with fluorescently labeled ph-Fab libraries. (A) Depending on the growth/display propensities of higher affinity ph- Fabs, iterative rounds of biopanning result in different relative populations of ph-Fab binders. (B) Where growth/display levels are similar between ph-Fab library members, flow distributions show an increase in normalized MFI while the standard deviation is similar (SD). (C) Where high- affinity ph-Fabs exhibit inferior growth/display propensities, MFI increase is halted prematurely, and SD decreases as weaker binders are more prevalent. (D) Where high-affinity ph-Fabs also exhibit superior growth/display propensities, an increasing in MFI and an increase in SD is observed.
FIG. 10 depicts a BATCH categorization, ranking, and output scheme.
FIG. 11 depicts a BATCH algorithm scheme. The thresholds for “true Assoc-2 curve slope”, “true Assoc-2 R2”, “extrapolated Assoc- 1 slope”, and “Dissoc R2” were all determined by the values obtained from the control experiments of P1A4 spiked in PPE. The “Assoc-1 Total change” threshold was determined by the control experiment where 25 nM of P1A4 was spiked in PPE, and BIAS koff values were more than 2-fold less than the true value.
FIGS. 12A to 12C optimization of phage labeling with NHS-FITC and M13 phage. Experiments were performed in n=3 and data is presented as mean ± SD. (A) Washing optimizations show three washing steps are sufficient for complete depletion of non-reacted NHS- FITC. (B) Fluorescent signal is increased as FITC labelling concentration is increased in FITC labeling. (C) FITC labeling is saturated after 1 hour reaction time. FIGS. 13A to 13B simple guide to ranking flagged clones from BIAS screens. (A) For candidates that exhibit comparable Assoc-2 slopes but show different BIAS kOffS, the clone with lower koff is the more promising binder. (B) For candidates that are flagged to not fully saturate or show very low expression levels, Assoc-2 curves are better indications of binding affinity than BIAS koft. Roughly, lower true Assoc-2 slopes indicate higher affinity.
FIGS. 14A to 14E flow cytometry analysis of fluorescent labeled phage displayed Fabs (ph-Fabs) bound to ARS1620-V7 immobilized beads. (A) ph-Fabs bound to ARS1620-V7-beads with varying incubation times. ph-PlA4 and ph-P2B2 were incubated for a total of 1 hour and 3 hours each. An increase in fluorescent intensity is observed beyond the 1-hour mark, indicating that the binding is not fully saturated even after 1 hour of incubation. (B) Flow cytometry analysis of ph-Fabs of varying affinities bound to ARS1620-V7-beads. Fluorescent distributions of beads bound to ph-P2B2, ph-PlH6, ph-PICl and ph-P2Fl 1 show MFI trends that agree with the ph-Fab affinities. (C) Flow cytometry analysis of increasing ph-PlA4 concentration shows an increase in normalized MFI with an observable shift from flow cytometry starting at 10 percent ph-Pl A4. (D) Overlay of flow cytometry analysis of ph-P2B2, ph-PlH6, and a ph-P2B2, ph-PlH6 mix (90%/10%) bound to ARS1720-V7-beads. (E) Flow cytometry analysis of fluorescein isothiocyanate (FITC) and Alexa Fluor 647 (AF647) labeled mixture of ph-P2B2 and ph-PlH6. Pure ph-P2B2 labeled with both FITC and AF647 was used as a control. Histograms of the dot blot distributions are also shown.
FIGS. 15 A to 15C rare antibody phage isolation and discrimination (RAPID) biopanning of Gaq. (A) RAPID flow cytometry of Rounds 1-6. Round 5 shows the highest normalized median fluorescent intensity (NMFI) of 2.36. (B) Overlay of RAPID flow cytometry of round 4 - 6 and a no phage control. An increase in standard deviation (SD) of the fluorescent distribution is observed in round 5. (C) Biolayer interferometry antibody screen (BIAS) kOff of candidate hits. Blue data points represent identical values, while red data points also represent identical values.
FIGS. 16A to 16D rare antibody phage isolation and discrimination (RAPID) biopanning of cyclic STAT3 decoy (CS3D). (A) RAPID flow cytometry of biopanning campaign rounds 1-5. No significant increase in normalized median fluorescent intensity (NMFI) is observed. (B) Zoom in of (A) populations where FITC- A >103. Maximum NMFI is exhibited in round 4. (C) Overlay of RAPID flow cytometry of round 4 (red, unfilled) and the original library control (grey, filled). (D) Biolayer interferometry antibody screen (BIAS) koffS of the top 4% and the bottom 96% of round 4 randomly chosen clones. The top ~4% sorted population contains ~2-fold lower BIAS koffS suggesting more promising binders exist in the sorted versus unsorted population of phage.
DEFINITIONS
“Derived from” in the context of an amino acid sequence or polynucleotide sequence is meant to indicate that the polypeptide or nucleic acid has a sequence that is based on that of a reference polypeptide or nucleic acid, and is not meant to be limiting as to the source or method in which the protein or nucleic acid is made.
An “anti-CS3D antibody” refers to an antibody that binds CS3D. An anti-CS3D antibody may bind to CS3D with a KD less than about 10'7 M, less than about 10’8, less than about 10'9, less than about 1O“10, less than about 10’11, or less than about 10’12 or less. In certain embodiments, “high affinity” antibodies have a KD of 100 nM or less.
The terms "polypeptide", "peptide", and "protein" are used interchangeably herein to designate a linear series of amino acid residues connected one to the other by peptide bonds between the alpha-amino and carboxy groups of adjacent residues. The amino acid residues are usually in the natural "L" isomeric form. However, residues in the "D" isomeric form can be substituted for any L-amino acid residue, as long as the desired functional property is retained by the polypeptide. In addition, the amino acids, in addition to the 20 "standard" amino acids, include modified and unusual amino acids, which include, but are not limited to those listed in 37 CFR (§ 1.822(b)(4)). Furthermore, it should be noted that a dash at the beginning or end of an amino acid residue sequence indicates either a peptide bond to a further sequence of one or more amino acid residues or a covalent bond to a carboxyl or hydroxyl end group. However, the absence of a dash should not be taken to mean that such peptide bonds or covalent bond to a carboxyl or hydroxyl end group is not present, as it is conventional in representation of amino acid sequences to omit such.
The term “antibody” (also used interchangeably with “immunoglobulin”) encompasses polyclonal and monoclonal antibody prepar ations where the antibody may be of any class of interest (e.g., IgM, IgG, and subclasses thereof), as well as preparations including hybrid antibodies, altered antibodies, F(ab')2 fragments, F(ab) molecules, Fv fragments, scFv fragments, single chain antibodies, single domain antibodies, chimeric antibodies, humanized antibodies, and functional fragments thereof which exhibit binding properties of the parent antibody molecule. The antibodies may be conjugated to other moieties, such as, labels, and/or may be bound to a support (c.g., a solid support), such as a polystyrene plate or bead, test strip, and the like.
Immunoglobulin polypeptides include the kappa and lambda light chains and the alpha, gamma (IgGi, IgGz, IgG s. IgG4), delta, epsilon and mu heavy chains or equivalents in other species. Full-length immunoglobulin “light chains” (usually of about 25 kDa or about 214 amino acids) include a variable region of about 110 amino acids at the NHi-terminus and a kappa or lambda constant region at the COOH-terminus. Full-length immunoglobulin “heavy chains” (of about 50 kDa or about 446 amino acids), similarly include a variable region (of about 116 amino acids) and one of the aforementioned heavy chain constant regions, e.g., gamma (of about 330 amino acids).
An immunoglobulin light or heavy chain variable region is composed of a “framework” region (FR) interrupted by three hypervariable regions, also called “complementarity determining regions” or “CDRs”. The extent of the framework region and CDRs have been defined (see, “Sequences of Proteins of Immunological Interest,” E. Kabat et al., U.S. Department of Health and Human Services, (1991) and Lefranc et al. IMGT, the international ImMunoGeneTics information system®. Nucl. Acids Res., (2005) 33:D593-D597)). A detailed discussion of the IMGTS system, including how the IMGTS system was formulated and how it compares to other systems, is provided on the World Wide Web at imgt.cines.fi7 textes/ IMGTScientificChart/ Numbering/ IMGTnumberingsTable.html. The sequences of the framework regions of different light or heavy chains are relatively conserved within a species. The framework region of an antibody, that is the combined framework regions of the constituent light and heavy chains, serves to position and align the CDRs. The CDRs are primarily responsible for binding to an epitope of an antigen. All CDRs and framework provided by the present disclosure are defined according to Kabat et al, supra, unless otherwise indicated. The three light chain CDRs, as used herein, are also referred to as “LCDR1”, “LCDR2”, and “LCDR3”. The three heavy chain variable CDRs, as used herein, are also referred to as “HCDR1”, “HCDR2”, and “HCDR3”.
An "antibody" thus encompasses a protein having one or more polypeptides that can be genetically encodable, e.g., by immunoglobulin genes or fragments of immunoglobulin genes. The recognized immunoglobulin genes include the kappa, lambda, alpha, gamma, delta, epsilon and mu constant region genes, as well as myriad immunoglobulin variable region genes. Light chains arc classified as cither kappa or lambda. Heavy chains arc classified as gamma, mu, alpha, delta, or epsilon, which in turn define the immunoglobulin classes, IgG, IgM, IgA, IgD and IgE, respectively.
A typical immunoglobulin (antibody) structural unit is known to include a tetramer. Each tetramer is composed of two identical pairs of polypeptide chains, each pair having one "light" (about 25 kD) and one "heavy" chain (about 50-70 kD). The N-terminus of each chain defines a variable region of about 100 to 110 or more amino acids primarily responsible for antigen recognition. The terms variable light chain (VL) and variable heavy chain (Vn) refer to these light and heavy chains respectively.
Antibodies encompass intact immunoglobulins as well as a number of well characterized fragments produced by digestion with various peptidases. Thus, for example, pepsin digests an antibody below the disulfide linkages in the hinge region to produce F(ab)'2, a dimer of Fab which itself is a light chain joined to VH-CH1 by a disulfide bond. The F(ab)'2 may be reduced under mild conditions to break the disulfide linkage in the hinge region thereby converting the (Fab')2 dimer into an Fab' monomer. The Fab' monomer is essentially an Fab with part of the hinge region (see, Fundamental Immunology, W.E. Paul, ed., Raven Press, N.Y. (1993), for a more detailed description of other antibody fragments). While various antibody fragments are defined in terms of the digestion of an intact antibody, one of skill will appreciate that such Fab' fragments may be synthesized de novo either chemically or by utilizing recombinant DNA methodology. Thus, the term antibody, as used herein also includes antibody fragments either produced by the modification of whole antibodies or synthesized de novo using recombinant DNA methodologies, including, but are not limited to, Fab'2, IgG, IgM, IgA, Fv, scFv, dAb, nanobodies (e.g., VH domain antibodies), unibodies, and diabodies.
Antibodies and fragments of the present disclosure encompass those that are bispecific. Bispecific antibodies or fragments can be of several configurations. For example, bispecific antibodies may resemble single antibodies (or antibody fragments) but have two different antigen binding sites (variable regions). Bispecific antibodies may be produced by chemical techniques (Kranz et al. (1981) Proc. Natl. Acad. Sci., USA, 78: 5807), by "polydoma" techniques (see, e.g., U.S. Pat. No. 4,474,893), or by recombinant DNA techniques. Bispecific antibodies may have binding specificities for at least two different epitopes, at least one of which is an epitope of CS3D.
An "antigen-binding site" or "binding portion" refers to the pail of an immunoglobulin molecule that participates in antigen binding. The antigen binding site is formed by amino acid residues of the N-terminal variable ("V") regions of the heavy ("H") and light ("L") chains. Three highly divergent stretches within the V regions of the heavy and light chains are referred to as "hypervariable regions" which are interposed between more conserved flanking stretches known as "framework regions" or "FRs". Thus, the term "FR" refers to amino acid sequences that are naturally found between and adjacent to hypervariable regions in immunoglobulins. In an antibody molecule, the three hypervariable regions of a light chain and the three hypervariable regions of a heavy chain are disposed relative to each other in three-dimensional space to form an antigen binding "surface". This surface mediates recognition and binding of the target antigen. The three hypervariable regions of each of the heavy and light chains are referred to as "complementarity determining regions" or "CDRs" and are characterized, for example by Kabat et al. Sequences of proteins of immunological interest, 4th ed. U.S. Dept. Health and Human Services, Public Health Services, Bethesda, MD (1987).
As used herein, the terms "immunological binding" and "immunological binding properties" refer to the non-covalent interactions of the type which occur between an immunoglobulin molecule and an antigen for which the immunoglobulin is specific. The strength or affinity of immunological binding interactions can be expressed in terms of the dissociation constant (KD) of the interaction, wherein a smaller KD represents a greater affinity. Immunological binding properties of selected polypeptides can be quantified using methods well known in the art. One such method entails measuring the rates of antigen binding site/antigen complex formation and dissociation, wherein those rates depend on the concentrations of the complex partners, the affinity of the interaction, and on geometric parameters that equally influence the rate in both directions. Thus, both the “on rate constant” (kon) and the "off rate constant" (koff) can be determined by calculation of the concentrations and the actual rates of association and dissociation. The ratio of koff/kOn enables cancellation of all parameters not related to affinity and is thus equal to the equilibrium dissociation constant KD (see, generally, Davies et. al., Ann. Rev. Biochem. 1990, 59: 439-15 473). An “epitope" is a site on an antigen (e.g., CS3D) to which an antibody hinds. “Antigen” and “target of interest” arc used interchangeably.
“Isolated” refers to an entity of interest that is in an environment different from that in which the compound may naturally occur or is initially produced in. An “isolated” compound or compounds (e.g., an “isolated” antibody or “isolated” antibodies) are separated from all or some of the components that accompany it and may be substantially enriched, e.g., may be purified so that the compound is at least about 70% pure, at least about 80% pure, at least about 90% pure, at least about 95% pure, at least about 98% pure, at least about 99%, or greater than 99% pure, or free of impurities, contaminants, and/or components other than the compound. “Isolated” also refers to the state of a compound separated from all or some of the components that accompany it during manufacture (e.g., chemical synthesis, recombinant expression, culture medium, and the like).
A single chain Fv ("scFv") polypeptide is a covalently linked VH::VL heterodimer which may be expressed from a nucleic acid including VH- and VL- encoding sequences either joined directly or joined by a peptide-encoding linker (Huston, et al. (1988) Proc. Nat. Acad. Sci. USA, 85: 5879-5883). A number of structures are available for converting the light and heavy polypeptide chains from an antibody V region into an scFv molecule which will fold into a three dimensional structure substantially similar to the structure of an antigen-binding site. See, e.g. U.S. Patent Nos. 5, 091,513 and 5,132,405 and 4,956,778.
Recombinant design methods may be used to develop suitable chemical structures (linkers) for converting two heavy and light polypeptide chains from an antibody variable region into a scFv molecule which will fold into a three-dimensional structure that is substantially similar to native antibody structure.
Design criteria include determination of the appropriate length to span the distance between the C-terminal of one chain and the N-terminal of the other, wherein the linker is generally formed from small hydrophilic amino acid residues that do not tend to coil or form secondary structures. Such methods have been described in the art. See, e.g., U.S. Patent Nos. 5,091,513 and 5,132,405 to Huston et al.', and U.S. Patent No. 4,946,778 to Ladner et al.
In this regard, the first general step of linker design involves identification of plausible sites to be linked. Appropriate linkage sites on each of the VH and VL polypeptide domains include those which will result in the minimum loss of residues from the polypeptide domains, and which will necessitate a linker including a minimum number of residues consistent with the need for molecule stability. A pair of sites defines a "gap" to be linked. Linkers connecting the C-terminus of one domain to the N-terminus of the next generally include hydrophilic amino acids which assume an unstructured configuration in physiological solutions and may be free of residues having large side groups which might interfere with proper folding of the VH and VL chains. Thus, suitable linkers generally include polypeptide chains of alternating sets of glycine and serine residues, and may include glutamic acid and lysine residues inserted to enhance solubility. One particular linker has the amino acid sequence (Gly4Ser)3 (SEQ ID NO:2). Another example of a suitable linker is a linker that has the amino acid sequence including 2 or 3 repeats of [(Ser)4Gly] (SEQ ID NO:3), such as [(Ser)4Gly]3 (SEQ ID NO:4), and the like. Nucleotide sequences encoding such linker moieties can be readily provided using various oligonucleotide synthesis techniques known in the art (see, e.g., Sambrook, supra.).
The phrase "specifically binds to" or "specifically immunoreactive with", when referring to an antibody that specifically binds to an antigen refers to a binding reaction which is determinative of the presence of the antigen in the presence of a heterogeneous population of other molecules, proteins, DNA, and other biologies. Thus, under designated immunoassay conditions, the specified antibodies bind to a particular antigen and do not bind in a significant amount to other molecules present in the sample. Specific binding to an antigen under such conditions may require an antibody that is selected for its specificity for the antigen. A variety of immunoassay formats may be used to select antibodies specifically immunoreactive with a particular antigen. For example, solid-phase enzyme-linked immunosorbent assay (ELISA) immunoassays are routinely used to select monoclonal antibodies specifically immunoreactive with an antigen. See Harlow and Lane (1988) Antibodies, A Laboratory Manual, Cold Spring Harbor Publications, New York, for a description of immunoassay formats and conditions that can be used to determine specific immunoreactivity.
The term "conservative substitution" is used in reference to proteins or peptides to reflect amino acid substitutions that do not substantially alter the activity (specificity or binding affinity) of the molecule. Typically, conservative amino acid substitutions involve substituting one amino acid for another amino acid with similar chemical properties (e.g. charge or hydrophobicity). The following six groups each contain amino acids that are typical conservative substitutions for one another: 1) Alanine (A), Serine (S), Threonine (T); 2) Aspartic acid (D), Glutamic acid (E); 3) Asparagine (N), Glutamine (Q); 4) Arginine (R), Lysine (K); 5) Isoleucine (I), Leucine (L), Methionine (M), Valine (V); and 6) Phenylalanine (F), Tyrosine (Y), Tryptophan (W).
DETAILED DESCRIPTION
The present disclosure provides methods and systems for identifying rare high-affinity antibodies against challenging targets using a recombinant phage displayed library. In the disclosure, both profiling methods for monitoring the enrichment of a biopanning campaign as well as screening methods for identifying, ranking, and prioritizing candidate binding antibodies are described. In addition, the profiling and screening methods are coupled together to form a single identification pipeline, wherein the disclosed profiling techniques are used to selectively isolate the most promising population of binding antibodies and the disclosed screening techniques subsequently perform screening in a discriminatory fashion. Also provided are systems for use in practicing methods of the invention.
Before the present invention is described in greater detail, it is to be understood that this invention is not limited to particular embodiments described, as such may, of course, vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to be limiting, since the scope of the present invention will be limited only by the appended claims.
Where a range of values is provided, it is understood that each intervening value, to the tenth of the unit of the lower limit unless the context clearly dictates otherwise, between the upper and lower limit of that range and any other stated or intervening value in that stated range, is encompassed within the invention. The upper and lower limits of these smaller ranges may independently be included in the smaller ranges and are also encompassed within the invention, subject to any specifically excluded limit in the stated range. Where the stated range includes one or both of the limits, ranges excluding either or both of those included limits are also included in the invention.
Certain ranges are presented herein with numerical values being preceded by the term "about." The term "about" is used herein to provide literal support for the exact number that it precedes, as well as a number that is near to or approximately the number that the term precedes. In determining whether a number is near to or approximately a specifically recited number, the near or approximating unrecited number may be a number which, in the context in which it is presented, provides the substantial equivalent of the specifically recited number.
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although any methods and materials similar or equivalent to those described herein can also be used in the practice or testing of the present invention, representative illustrative methods and materials are now described.
All publications and patents cited in this specification are herein incorporated by reference as if each individual publication or patent were specifically and individually indicated to be incorporated by reference and are incorporated herein by reference to disclose and describe the methods and/or materials in connection with which the publications are cited. The citation of any publication is for its disclosure prior to the filing date and should not be construed as an admission that the present invention is not entitled to antedate such publication by virtue of prior invention. Further, the dates of publication provided may be different from the actual publication dates, which may need to be independently confirmed.
It is noted that, as used herein and in the appended claims, the singular forms “a”, “an”, and “the” include plural referents unless the context clearly dictates otherwise. It is further noted that the claims may be drafted to exclude any optional element. As such, this statement is intended to serve as antecedent basis for use of such exclusive terminology as “solely,” “only” and the like in connection with the recitation of claim elements, or use of a “negative” limitation.
As will be apparent to those of skill in the art upon reading this disclosure, each of the individual embodiments described and illustrated herein has discrete components and features which may be readily separated from or combined with the features of any one of the other several embodiments without departing from the scope or spirit of the present invention. Any recited method can be carried out in the order of events recited or in any other order which is logically possible.
While the apparatus and method has or will be described for the sake of grammatical fluidity with functional explanations, it is to be expressly understood that the claims, unless expressly formulated under 35 U.S.C. §112, are not to be construed as necessarily limited in any way by the construction of "means" or "steps" limitations, but are to be accorded the full scope of the meaning and equivalents of the definition provided by the claims under the judicial doctrine of equivalents, and in the case where the claims are expressly formulated under 35 U.S.C. §1 12 are to be accorded full statutory equivalents under 35 U.S.C. §112.
METHODS
As summarized above, methods for identifying rare high-affinity antibodies against challenging targets using a recombinant phage displayed library are provided. In the disclosure, both profiling methods for monitoring the enrichment of a biopanning campaign as well as screening methods for identifying, ranking, and prioritizing candidate binding antibodies are described. In addition, the profiling and screening methods are coupled together to form a single identification pipeline, wherein the disclosed profiling techniques are used to selectively isolate the most promising population of binding antibodies and the disclosed screening techniques subsequently perform screening in a discriminatory fashion. Also provided are systems for use in practicing methods of the invention.
Phage Display Biopanning
As described above, embodiments of the methods include monitoring a biopanning campaign by generating a profile for each round of the campaign. Biopanning techniques, in accordance with embodiments of the methods, may vary and may include, but are not limited to, any one of the phage display biopanning techniques discussed below or any standard phage display biopanning technique, as well as combinations thereof, as is known in the art.
In some embodiments, phage display biopanning includes one or more of the steps of: obtaining or generating a phage display library including a multitude of different antibodies (e.g., a multitude of different fragment antigen-binding regions (Fabs)) displayed on phages; contacting the phage display library with an antigen or target of interest in order to bind phages displaying antibodies with affinity to the antigen or target of interest; washing away or separating out unbound phages from phages bound to the target of interest; eluting phages bound to the target of interest; and amplifying the eluted phages and antibodies displayed thereon in order to perform an additional round of biopanning (e.g., by infecting bacteria cells with the eluted phages). In some embodiments, the biopanning steps are repeated in order to perform multiple rounds of biopanning. In other words, each biopanning step is repeated for each additional round of biopanning performed excluding, in some instances, the final amplification step from the final round of biopanning. In some embodiments, two or more rounds of biopanning are performed such as, c.g., three or more rounds of biopanning, or four or more, or five or more, or ten or more, or fifteen or more, or twenty or more, or fifty or more, or one hundred or more. By “biopanning campaign” is meant one or more connected rounds of biopanning, connected rounds being rounds wherein the final amplification step of a first round is used to generate the phage display library of a subsequent round of biopanning.
As discussed above, phage display biopanning may include the step of obtaining or generating a phage display library including a multitude of different antibodies (e.g., a multitude of different Fabs) displayed on phages. The antibodies may be any of type antibody, or antibody fragment, as discussed above including, e.g., Fabs, Fvs, scFvs, VH domain antibodies, or nanobodies. The methods of obtaining or generating a phage display library may vary. For the subsequent biopanning rounds performed after an initial round of biopanning, the phage display library may be generated by amplifying the eluted phages of a previous round of biopanning (e.g., by infecting bacteria with the phages), as discussed in greater detail below. For the initial round of biopanning, the antibody library (e.g., Fab library) displayed on phages may be obtained from an already existing library or may be generated using any known antibody engineering technique. In embodiments where the antibody library is obtained from an existing library, the library may be a human antibody library such as, e.g., a human naive B-cell Fab library. The obtained antibody library may then be used to generate the phage display library by inserting each desired antibody gene or gene fragment (i.e., a nucleic acid sequence encoding the gene or gene fragment) into the genome of a phage (i.e., bacteriophage) in a manner such that the antibodies are displayed on the surface of the phage virions. In embodiments where the antibody library is generated using antibody engineering techniques, the antibody library may be generated, e.g., by altering antibodies known to have affinity to the target of interest (using, e.g., techniques such as site saturation mutagenesis, error prone PCR, or DNA shuffling), or through in silica techniques such as by applying machine learning techniques to discover novel antibodies. The generated antibody library may then be used to generate the phage display library as discussed above.
In some embodiments, the phage display library may include a multitude of different antibodies such as, e.g., 104 different antibodies or more. In some cases, the phage display library may include 105 different antibodies or more, or 106 or more, or 107 or more, or 108 or more, or 109 or more, or IO10 or more, or 1011 or more, or 1012 or more. In some embodiments, the phage display library may include a range of 109 to 1012 different antibodies, or IO10 to 1011 different antibodies.
The phages of the phage display library may include any phage capable of displaying antibodies of the antibody library on its surface and infecting a bacteria in laboratory reproducible conditions in order to perform the amplification step of phage biopanning. In some embodiments, the phages may include the M13 phage, the T4 phage, the T7 phage, and/or the /. phage. In some instances, the phages of the phage display library are M13 phages.
As discussed above, phage display biopanning may include the step of contacting the phage display library with an antigen or target of interest in order to bind phages displaying antibodies with affinity to the antigen or target of interest. The antigen or target of interest, and the technique used to contact the phage display library with the target of interest, may vary as desired.
In some embodiments, the antigen or target of interest may include protein antigens or non-protein antigens. In embodiments wherein the antigen is a non-protein antigen, the antigen may include a nucleic acid such as, e.g., a DNA or RNA molecule. In some cases, the nucleic acid has a distinct structural motif such as, e.g., a helix-tum-helix motif, a hairpin motif, a kink, a three- or four-way junction, etc. In other cases, the nucleic acid may not have a distinct structural motif. In some embodiments, the antigen or target of interest may have low antigenicity or may have conformational heterogeneity (e.g., the antigen/target of interest may be conformationally dynamic). For example, the antigen or target of interest may have low thermal stability, be conformationally diverse, have low expression, or have low solubility.
The technique used to contact the phage display library with the antigen or target of interest may include any suitable techniques, or combinations thereof, known in the art. In some embodiments, the target of interest may be affixed to a stable surface such as, e.g., the walls of a microtiter plate. In these cases, the phage display library may then be introduced into the microtiter plate. In some cases, the target of interest may be affixed to free floating (e.g., in liquid media) particles such as, e.g., a plurality of beads. In some instances, multiple copies of the target of interest may be affixed to each one of the plurality of beads. For example, each bead may include two or more copies of the target of interest, such as three or more, or five or more, or ten or more, or twenty or more, or 100 or more. In some embodiments, the beads may be streptavidin beads. Tn some embodiments, the beads may be magnetic beads. In embodiments where the target of interest is affixed to free floating particles (c.g., beads), the phage display library may be contacted with the antigen or target of interest by introducing the free-floating particles into the same liquid media as the phage display library.
As discussed above, phage display biopanning may include the step of washing away or separating out unbound phages from phages bound to the target of interest. Any suitable buffer known in the art may be used to wash away or separate out unbound phages from phages bound to the target of interest. In embodiments where the target of interest is affixed to magnetic beads, the unbound phages may be separated from phages bound to the target of interest by applying a magnetic field to the magnetic beads.
As discussed above, phage display biopanning may include the step of eluting phages bound to the target of interest. Any suitable buffer known in the art may be used to elute phages bound to the target of interest. The elution step may subsequently be followed up by an amplification step wherein the eluted phages are amplified prior to a subsequent round of biopanning. The amplification step may occur by infecting bacteria cells (i.e., gram-negative bacteria cells) with the eluted phages in order to cause the bacteria to produce additional copies of the eluted phages. The bacteria used in the amplification step may include any bacteria capable of being infected by, and producing, phages of the phage display library in laboratory reproducible conditions. In some embodiments, the bacteria may be Escherichia coli bacteria.
In some embodiments, a subset of the eluted phages is obtained or stored for each round of biopanning performed in order to, e.g., generate a profile for each round of biopanning as discussed in greater detail below. In some instances, the subset of the eluted phages is obtained or stored for a round of biopanning after the amplification step is performed.
Profiling and Isolating Phages
As described above, embodiments of the methods include profiling techniques for identifying antibodies (e.g., a plurality of different antibodies) with affinity to a target of interest. In some cases, the profiling techniques are used to monitor a biopanning campaign (e.g., as described above) by generating a profile for each round of the campaign. Aspects of the profiling methods may include: conducting a first round of biopanning including isolating phages displaying antibodies that bind to the target of interest and fluorescently labeling the phages to generate a first labeled population of phages; conducting a second round of biopanning including isolating phages displaying antibodies that bind to the target of interest and fluorescently labeling the phages to generate a second labeled population of phages; separately contacting the first and the second labeled population of phages with the target of interest immobilized on beads to produce bead-phage complexes; analyzing the complexes using flow cytometry to generate a first fluorescence profile of the complexes produced from the first labeled population of phages and a second fluorescence profile of the second labeled population of phages; and using the first and second profiles to identify antibodies with affinity to the target of interest.
In some embodiments, two rounds of biopanning are conducted to generate two fluorescence profiles as described above. In some cases, three or more rounds of biopanning are conducted to generate three or more fluorescence profiles, such as four or more rounds of biopanning, or five or more rounds, or ten or more rounds, or twenty or more rounds.
The two or more populations of phages (i.e., corresponding to the two or more rounds of biopanning) may be fluorescently labeled through any number of suitable means. For example, any method of fluorescently labeling a particle known in the art that is capable of labeling the phages with minimal variability between each labeled phage (e.g., such that each phage includes roughly the same number of fluorescent labels) without disrupting interactions between the displayed antibodies and the beads including the target of interest may be used. For example, in embodiments where the phage is an M13 phage, the Ml 3 phage may be fluorescently labeled through a chemical reaction that efficiently labels the N-termini and lysine residues of the pVIII coat proteins of the M13 phage.
The fluorescent label may include a polymeric or a non-polymeric dye or fluorophore. In some embodiments, the fluorescent label may include one or more of: 4-acetamido-4’- isothiocyanatostilbene-2,2’disulfonic acid; acridine and derivatives such as acridine, acridine orange, acridine yellow, acridine red, and acridine isothiocyanate; 5-(2’- aminoethyl)aminonaphthalene- 1 -sulfonic acid (EDANS); 4-amino-N-[3- vinylsulfonyl)phenyl]naphthalimide-3,5 disulfonate (Lucifer Yellow VS); N-(4-anilino-l- naphthyl)maleimide; anthranilamide; Brilliant Yellow; coumarin and derivatives such as coumarin, 7-amino-4-methylcoumarin (AMC, Coumarin 120), 7-amino-4- trifluoromethylcouluarin (Coumaran 151); cyanine and derivatives such as cyanosine, Cy3, Cy3.5, Cy5, Cy5.5, and Cy7; 4’,6-diaminidino-2-phenylindole (DAPI); 5’, 5”- dibromopyrogallol-sulfonephthalein (Bromopyrogallol Red); 7-diethylamino-3-(4’- isothiocyanatophcnyl)-4-mcthylcoumarin; dicthylaminocoumarin; dicthylcnctriaminc pentaacetate; 4,4’ -diisothiocyanatodihydro-stilbene-2, 2’ -disulfonic acid; 4,4’- diisothiocyanatostilbene-2,2’-disulfonic acid; 5-[dimethylamino]naphthalene-l-sulfonyl chloride (DNS, dansyl chloride); 4-(4’-dimethylaminophenylazo)benzoic acid (DABCYL); 4- dimethylaminophenylazophenyl-4’-isothiocyanate (DABITC); eosin and derivatives such as eosin and eosin isothiocyanate; erythrosin and derivatives such as erythrosin B and erythrosin isothiocyanate; ethidium; fluorescein and derivatives such as 5-carboxy fluorescein (FAM), 5- (4,6-dichlorotriazin-2-yl)aminofluorescein (DTAF), 2’7’-dimethoxy-4’5’-dichloro-6- carboxyfluorescein (JOE), fluorescein isothiocyanate (FITC), fluorescein chlorotriazinyl, naphthofluorescein, and QFITC (XRITC); fluorescamine; IR144; IR1446; Green Fluorescent Protein (GFP); Reef Coral Fluorescent Protein (RCFP); Lissamine™; Lissamine rhodamine, Lucifer yellow; Malachite Green isothiocyanate; 4-methylumbelliferone; ortho cresolphthalein; nitrotyrosine; pararosaniline; Nile Red; Oregon Green; Phenol Red; B -phycoerythrin (PE); o- phthaldialdehyde; pyrene and derivatives such as pyrene, pyrene butyrate and succinimidyl 1- pyrene butyrate; Reactive Red 4 (Cibacron™ Brilliant Red 3B-A); rhodamine and derivatives such as 6-carboxy-X-rhodamine (ROX), 6-carboxyrhodamine (R6G), 4,7 -dichlororhodamine lissamine, rhodamine B sulfonyl chloride, rhodamine (Rhod), rhodamine B, rhodamine 123, rhodamine X isothiocyanate, sulforhodamine B, sulforhodamine 101, sulfonyl chloride derivative of sulforhodamine 101 (Texas Red), N,N,N’,N’-tetramethyl-6-carboxyrhodamine (TAMRA), tetramethyl rhodamine, and tetramethyl rhodamine isothiocyanate (TRITC); riboflavin; rosolic acid and terbium chelate derivatives; xanthene; carotenoid-protein complexes, such as peridinin-chlorophyll proteins (PerCP); allophycocyanin (APC); Alexa Fluor™ (AF) dyes such as AF350, AF546, AF647, and AF790; or combinations thereof. In some embodiments, the fluorescent label may be FITC or an AF dye (e.g., AF647). In some cases, N- hydroxysuccinimide (NHS)-ester labeling reagents may be used to label the phages. In these instances, NHS-fluorescein, NHS-FITC, and/or NHS-AF647 may be used to label the phages.
As discussed above, embodiments of the profiling methods may include the step of contacting the fluorescently labeled phages with beads including the target of interest to produce bead-phage complexes. In these instances, each bead may include multiple copies of the target of interest such that multiple fluorescently labeled phages may bind to each bead (such as, e.g., two or more copies of the target of interest, or five or more, or ten or more, or twenty or more, or fifty or more). In some embodiments, each population of fluorescently labeled phages (c.g., corresponding to each of the two or more rounds of biopanning as discussed above) are separately contacted (e.g., in separate aliquots of a liquid) with the target of interest immobilized on/affixed to beads to produce separate populations of bead-phage complexes. Any free floating (e.g., in liquid media) particles or beads capable of being affixed to one or more copies of the target of interest or antigen may be used (i.e., wherein the target of interest or antigen is immobilized on the particles or beads). In some embodiments, the beads may be streptavidin beads. In these cases, the antigen or target of interest may be biotinylated in order to affix the antigen to (i.e., immobilize the antigen on) the streptavidin beads. The streptavidin beads including the antigen or target of interest may then be introduced into the same liquid media (e.g., buffer or solution) as the labeled phages.
In some embodiments, the fluorescently labeled phages are incubated with the beads including (e.g., affixed to) the target of interest for a time period sufficient to produce beadphage complexes. In these instances, the incubation time period may be one minute or more, or five minutes or more, or twenty minutes or more, or an hour or more, or two hours or more. After the bead-phage complexes have been produced, the complexes may be washed multiple times and/or passed through a strainer in order to separate the complexes from unbound phages.
As discussed above, embodiments of the profiling methods may include the step of analyzing the complexes using flow cytometry to generate a fluorescence profile for each population of fluorescently labeled phage/bead complexes (e.g., corresponding to each of the two or more rounds of biopanning fluorescently labeled and contacted with the target of interest immobilized on beads, as described above). Suitable flow cytometry systems may include, but are not limited to, those described in Ormerod (ed.), Flow Cytometry: A Practical Approach, Oxford Univ. Press (1997); Jaroszeski et al. (eds.), Flow Cytometry Protocols, Methods in Molecular Biology No. 91, Humana Press (1997); Practical Flow Cytometry, 3rd ed., Wiley-Liss (1995); Virgo, et al. (2012) Ann Clin Biochem. Jan;49(pt 1): 17- 28; Linden, et. al., Semin Throm Hemost. 2004 Oct;30(5):502-l l; Alison, et al. J Pathol, 2010 Dec; 222(4):335-344; and Herbig, et al. (2007) Crit Rev Ther Drug Carrier Syst. 24(3):203-255; the disclosures of which are incorporated herein by reference. In certain instances, flow cytometry systems of interest include a BD Biosciences FACSCanto™ II flow cytometer, a BD Accuri™ flow cytometer, a BD Biosciences FACSCount™ cell sorter, a BD Biosciences Aria™ cell sorter and/or a Beckman Cytoflcx Analyzer, or the like. In some embodiments, the complexes may be gated using forward scatter (FSC) and/or side scatter (SSC) parameters.
The fluorescence profile generated for each round of biopanning using flow cytometry may include any graph or chart having information associated with the binding affinity of antibodies of the phage display library to the target or interest and/or one or more other dominant factors that may have affected enrichment (e.g., growth rates of phage containing bacteria, antibody displaying propensities, etc.). In some embodiments, the profile may include a histogram representing the frequency of complexes having different fluorescence intensities. In these cases, the bin or channel sizes of the histogram may be adjusted depending on, e.g., the variability of the fluorescence intensities of the complexes being analyzed or the number of complexes being analyzed.
In some embodiments, the fluorescence profiles generated for each round of biopanning using flow cytometry may be used to identify antibodies with high affinity to the target or antigen of interest. In some cases, the identified high-affinity antibodies may be the most promising population of binding antibodies (i.e., the population having the highest affinity to the target of interest) of the two or more populations of phage display libraries (i.e., corresponding to the populations generated during the two or more rounds of biopanning as discussed above). In some cases, the identified high-affinity antibodies may be a subgroup/subpopulation of a population generated from one of the two or more rounds of biopanning. In some embodiments, one or more metrics may be calculated for each profile. In some embodiments, one or more metrics may be calculated for one or more components of each profile. For example, in some instances multiple subpopulations may be present in a profile, and one or more metrics may be calculated for each subpopulation. The one or more metrics may include metrics indicating the binding affinity of antibodies of the phage display library to the target or interest and/or metrics indicating the variability of a population of antibodies. For example, a median fluorescence intensity (MFI) and/or a measure of variability (e.g., a standard deviation [SD]) may be calculated for each fluorescence profile (or, e.g., each subpopulation of each profile) when the profile is, e.g., a histogram as described above. In some embodiments, a single MFI and SD metric may be calculated for a generated histogram when, e.g., the histogram is unimodal. In some embodiments, an MFI and SD metric may be calculated for each mode (i.e., subpopulation) of a generated histogram when, e.g., the generated histogram is multimodal. The number of modes of each histogram may be determined visually (e.g., by a user) or through any number of statistical techniques such as, e.g., using the Bayesian information criterion, Akiakie information criterion, Calinski-Harabasz criterion, etc.
In some embodiments, the calculated metrics of each fluorescence profile may be used in order to identify antibodies with high affinity to the target or antigen of interest. For example, in embodiments where the profiles include histograms (e.g., as described above), antibodies associated with (i.e., corresponding to) histograms (or, e.g., modes of the histograms) having an MFI above a predetermined threshold value may be identified as high affinity antibodies. In some cases, antibodies associated with the histograms (or, e.g., the modes of the histograms) having the highest MFIs may be identified as high affinity antibodies. In some cases, antibodies associated with the histograms (or, e.g., the modes of the histograms) having the highest SDs may be identified as high affinity antibodies when, e.g., the MFIs of the histograms are relatively similar. In some embodiments, the antibodies associated with the profile, or the component of the profile (e.g., a mode corresponding to a subpopulation), with the highest MFI and SD are identified as high affinity antibodies. In some embodiments, the identified high affinity antibodies are isolated for downstream processing including, e.g., bio-layer interferometry (BLI) screening techniques as described in greater detail below using fluorescence activated sorting.
In some embodiments, a whole round of biopanning may be isolated as, e.g., the most promising population of binding antibodies within the one or more populations of phage display libraries. In other embodiments, one or more subsets (e.g., modes corresponding to subpopulations) of one or more of the phage display libraries (i.e., corresponding to the one or more rounds of biopanning) may be isolated using the generated fluorescence profiles as discussed above. In these instances, fluorescence activated sorting may be employed to isolate a plurality of high affinity antibodies corresponding to one or more specific components (e.g., modes corresponding to subpopulations) of the generated profiles as discussed above. In these cases, the flow cytometric systems (i.e., as described above) may be configured to sort one or more of the bead-phage complexes. The term “sorting” is used herein in its conventional sense to refer to separating components (e.g., complexes) of the sample and in some instances delivering the separated components to one or more sample collection containers. In some embodiments, the complexes may be sorted based on a level of fluorescent phages bound to the beads, thereby separating complexes exhibiting a threshold level of fluorescence from complexes exhibiting fluorescence below the threshold level. High affinity antibodies (c.g., the most promising population of binding antibodies within the one or more populations of phage display libraries) may then be separated from the complexes exhibiting the threshold level of fluorescence.
In some embodiments, the identified antibodies with affinity to the target of interest (as described above) may be screened for false positives, negatives, and hits using the bio-layer interferometry (BLI) screening method as discussed in greater detail below. In some instances, antibodies determined to be hits may be ranked using, e.g., a determined binding kinetic parameter, and the top ranked hits may be sequenced in order to identify one or more high- affinity antibodies against the target of interest or antigen.
Biolayer Interferometry Antibody Screening (BIAS)
As described above, embodiments of the methods include screening techniques for identifying, ranking, and prioritizing candidate binding antibodies. In some cases, the candidate binding antibodies are identified and, e.g., isolated for screening using the profiling methods for monitoring the enrichment of a biopanning campaign as discussed above. Aspects of the BLI screening methods for identifying antibodies with affinity to a target of interest may include: contacting a sensor tip including the target of interest immobilized thereon with a first aliquot of a solution including multiple copies of a first antibody; irradiating the sensor tip with an incident light and measuring a first reflected light, wherein a wavelength shift of the first reflected light compared to the incident light is indicative of binding of the first antibody to the target of interest immobilized on the sensor tip; contacting the sensor tip with a second aliquot of the solution including multiple copies of the first antibody, wherein the second aliquot further includes a second antibody that binds to the first antibody; and irradiating the sensor tip with the incident light and measuring a second reflected light, wherein a wavelength shift of the second reflected light compared to the incident light and/or the first reflected light is indicative of binding of the second antibody to the first antibody bound to the sensor tip, wherein presence of: (a) the wavelength shift of first reflected light compared to the incident light and (b) the wavelength shift of the second reflected light compared to the incident light and/or the first reflected light identifies the first antibody as having affinity to the target of interest. In some embodiments, the sensor tip is a bio-layer interferometry sensor tip. In some embodiments, the BLI sensor tip may include, e.g., a surface including one or more of a target or antigen of interest, a light source for interrogating the surface, and a detector for detecting light reflected off of the surface (e.g., and components affixed and/or bound to the surface). In some embodiments, the light source may be any light source capable of producing a white light such as, e.g., a broadband white light source. In some instances, the light source is a tungsten lamp or one or more LEDs. In some embodiments, the sensor tip is configured to measure interactions between the target of interest (i.e., on the surface of the sensor tip), the copies of an antibody of the plurality of antibodies (e.g., isolated using the profiling techniques as discussed above), and anti-tag binding antibodies (e.g., as discussed in greater detail below) by irradiating the surface including the target of interest with incident light and measuring the light reflected from the surface to determine the wavelength shift over time using the sensor tip.
As discussed above, embodiments of the screening methods may include the step of contacting a first aliquot of a solution including multiple copies of a first antibody of (e.g., one of the high affinity antibodies identified using the profiling techniques as discussed above) with a sensor tip including the target of interest. In some cases, when the first aliquot of the solution is contacted by the sensor tip including the target of interest (e.g., when the sensor top is dipped or inserted into the first aliquot of the solution) copies of the antibody may begin to bind or associate with the target of interest. In these instances, the measured wavelength of the light reflected off of the surface of the sensor tip including the target of interest may continuously shift (i.e., when compared to the incident light) as more copies of the antibody bind to the target of interest. In some cases, the wavelength shift of the reflected light compared to the incident light is indicative of binding of the first antibody to the target of interest immobilized on the sensor tip. In some embodiments, a curve is fit to the measurements of the wavelength shift (i.e., of the measured reflected light compared to the incident light) generated over the time the sensor is in the first aliquot of the solution. In these instances, the curve fit to the wavelength shift measurements may be a one phase exponential association curve. In some embodiments, the rate of change of wavelength shift over time (e.g., the slope of the fitted curve) may be determined for one or more timepoints corresponding to one or more measurements generated by the sensor tip while the sensor tip is in the first aliquot of the solution. For example, the rate of change of wavelength shift over time may be determined for the last measurement before the sensor tip is removed from the first aliquot of solution and inserted into the second aliquot of solution.
As discussed above, embodiments of the screening methods may include the step of contacting a second aliquot of solution with the sensor tip, wherein the second aliquot of the solution includes the copies of the first antibody and a second antibody (e.g., multiple copies of the second antibody) configured to bind to the first antibody. The second antibody configured to bind to the first antibody may include any antibody (e.g., obtained from any organism or laboratory technique) capable of binding to the first antibody. In some cases, each copy of the first antibody of the includes a tag. The tag may be any tag that is capable of being associated or affixed to each antibody and capable of being bound by a secondary antibody that does not disrupt interactions between the antibodies and the target of interest. In some embodiments, the tag is a myc-tag. In some embodiments, the second antibody (i.e., that binds to the first antibody) is configured to bind to the tag of the first antibody, and may include any antibody (e.g., obtained from any organism or laboratory technique) capable of binding to the tag associated or affixed to each copy of the first antibody as described above. In embodiments where the tag is the myc-tag, the second antibody configured to bind to the tag may include anti-myc antibodies (e.g., anti- Myc IgG).
In some cases, when the second aliquot of solution is contacted by the sensor tip including the target of interest (e.g., when the sensor tip is dipped or inserted into the second aliquot of the solution) first antibody copies may continue to bind or associate with the target of interest and the second antibody (e.g., one or more copies of the second antibody) may begin to bind or associate with copies of the first antibody (e.g., a tag of the first antibody copies). In these instances, the measured wavelength of the light reflected off of the surface of the sensor tip including the target of interest may continuously shift (e.g., when compared to the incident light and/or the first reflected light) as copies of the first antibody continue to bind to the target of interest and the second antibody binds to copies of the first antibody that are bound to the target of interest. In some cases, the wavelength shift of the reflected light when the sensor tip is in the second aliquot compared to the incident light and/or the reflected light when the sensor tip was in the first aliquot is indicative of binding of the first antibody to the target of interest immobilized on the sensor tip. In some embodiments, a curve is fit to the measurements of the wavelength shift (i.e., of the measured reflected light compared to the incident light) generated over the time the sensor is in the second aliquot of the solution. In these instances, the curve fit to the measurements may be a line. In some embodiments, the rate of change of wavelength shift over time (e.g., the slope of the fitted line) may be determined for the fitted line. In some embodiments, the curve fit for the first aliquot measurements may be extrapolated through the period of time of the second aliquot measurements. In these instances, the measurements generated while the sensor tip is in the second aliquot of solution may be subtracted by the extrapolated curve. In these cases, a line may be fit to the second aliquot measurements after subtraction and, e.g., the slope of the fitted line may be determined.
In some embodiments, the first aliquot of solution and the aliquot of solution may include (e.g., the solution of the first and second aliquots may be) crude periplasmic extract (PPE) produced from bacteria (such as, e.g., E. coli). In some cases, the bacteria used to produce the PPE is infected with phage in order to produce the identified high affinity antibodies of two or more rounds of biopanning as discussed above. In some instances, the first antibody as described above is one of the identified high affinity antibodies. In some cases, the concentration of the copies of the first antibody in both the first and second aliquot of the solution may be the same. In some instances, the concentration and makeup of the PPE in both the first and second aliquot of solution (e.g., the same solution may be the PPE).
In some embodiments, a difference in the rate of change of wavelength shift over time (i.e., of measured reflected light compared to incident light) between when the sensor tip is in contact with first aliquot and when the sensor tip is in contact with the second aliquot is used to identify whether the first antibody has affinity to the target of interest. In some embodiments, a higher positive rate of change of wavelength shift when the sensor tip is in the second aliquot compared to the first aliquot indicates the first antibody has affinity to the target of interest.
In some embodiments, a buffer is contacted with the sensor tip after the second aliquot of solution. In these instances, the copies of the first antibody may begin to dissociate from the target of interest while the sensor tip is inside of the buffer. In some embodiments, the buffer includes a lower concentration of copies of the first antibody (e.g., and the second antibody) than the first and/or second aliquot of solution. In some cases, the buffer includes no copies of the first and/or second antibody and may further include no PPE. In some embodiments, the wavelength of the measured light reflected off of the surface of the sensor tip including the target of interest may continuously shift (i.e., when compared to the incident light and the reflected light measured in the second aliquot of the solution) as copies of the first antibody dissociate from the target of interest. In some embodiments, a curve is fit to the measurements of the wavelength shift (i.e., of the measured reflected light compared to the incident light) generated over the time the sensor tip is in the buffer. In these instances, the curve fit to the measurements may be a one phase exponential decay curve. In some embodiments, the buffer includes a chaotropic agent. In other cases, the buffer does not include a chaotropic agent.
In some embodiments, a goodness of fit metric is calculated for each fitted curve (i.e., the curves fitted for the measurements taken while the sensor is in the first aliquot of solution, the second aliquot of solution, and the buffer). In these cases, the goodness of fit metric may be an r- squared value.
In some embodiments, the BLI screening is performed simultaneously or sequentially for a plurality of different antibodies (i.e., simultaneously, or sequentially on a plurality of solutions including different antibodies). In some embodiments, the plurality of antibodies is produced from a phage display library (i.e., of antibodies). In these cases, the phage display library may include phages having undergone two or more rounds of preselection (e.g., biopanning) for phages that bind to the target of interest. For example, the BLI screening may be performed for a plurality of high affinity antibodies (i.e., displayed on phages) identified using the profiling techniques as discussed above. In these embodiments, bacteria may be infected with the phage display library (e.g., phages expressing the high affinity antibodies) and the bacteria may be separated into single cells. The separated bacteria may then be used to grow bacterial colonies that each express a single type of antibody. In some embodiments, an extract may be prepared from the colonies to provide a plurality of solutions, wherein each solution includes multiple copies of a single type of antibody (i.e., multiple copies of the first antibody as described in the screening techniques above). The plurality of solutions may include first and second aliquots of each solution used for screening the antibody copies of each colony using the BLI screening techniques as described above (i.e., wherein the second aliquot includes a second antibody that binds to the first antibody [i.e., the single type of antibody produced by a bacteria colony]). In some cases, the screening is performed for the antibodies of phages produced from at least 10, at least 30, at least 100, at least 300, or more individual bacterial colonies (e.g., in parallel, wherein a plurality of sensor tips contact each first aliquot simultaneously followed by contacting each second aliquot simultaneously). In some embodiments, negatives of first antibody copies not binding to the target of interest, false positives of non-specific binding proteins binding to the target of interest, and/or hits of first antibody copies binding to the target of interest may be detected. In some embodiments, negatives and false positives are detected by determining if the slope of the line fit to the second aliquot measurements is below a predetermined threshold value and/or if the r- squared value of the line fit to the second aliquot measurements is below a predetermined threshold value. In these embodiments, false positives may be detected by determining if the r- squared value of the association curve fit to the first aliquot measurements meets or exceeds a predetermined threshold value (e.g., different than the threshold value used to distinguish negatives and false positives from hits). If the r-squared value does not meet or exceed the additional predetermined threshold value, the antibody copies may be determined to be negatives (i.e., non-binders).
In some embodiments, hits are detected by determining if the slope of the line fit to the second aliquot measurements meets of exceeds a predetermined threshold value and if the r- squared value of the line fit to the second aliquot measurements meets or exceeds a predetermined threshold value.
In some embodiments, whether or not the sensor tip is saturated is determined by calculating a slope of the association curve extrapolated through the period of time of the second aliquot measurements. In these instances, the sensor tip may be determined to be saturated if the extrapolated association curve meets or exceeds a predetermined threshold value.
In some embodiments, antibody copy expression is determined by calculating the total wavelength shift of the measurements occurring while the sensor tip is inside of the first aliquot of liquid. In these instances, the antibody copy expression may be determined to be low if the total wavelength shift of the first aliquot measurements is below a predetermined threshold value.
In some embodiments, a dissociation rate for the first antibody copies and the target of interest is predicted using the exponential decay curve fit to the buffer measurements. The dissociation rate may be determined to be inaccurate if the r-squared value of the exponential decay curve fit to the buffer measurements is below a predetermined threshold value. In some embodiments, a dissociation rate is predicted for a plurality of identified high affinity antibodies (i.e., identified using the profiling techniques as discussed above) and the predicted dissociation rates are used to rank each antibody of the plurality of antibodies. In some embodiments, the top ranked binding candidates arc sequenced in order to identify one or more high-affinity antibodies against the target of interest or antigen.
SYSTEMS
Aspects of the present disclosure further include systems, such as computer-controlled systems, for practicing embodiments of the above methods. Aspects of the systems may include: a processor including memory operably coupled to the processor wherein the memory includes instructions stored thereon, which when executed by the processor, cause the processor to: obtain raw bio-layer interferometry data, wherein the interferometry data includes sequential wavelength shift measurements resulting from interactions between a sensor tip including a target of interest, copies of an antibody, and tag binding antibodies; section the interferometry data into two consecutive segments; fit a one phase exponential association curve to raw data from the first segment, wherein the sequentially later end of the one phase exponential association curve includes a first slope; fit a line to the second segment data, wherein the fitted line includes a second slope; and compare the first slope to the second slope, wherein the antibody copies are identified as having affinity to a target of interest if the second slope is greater than the first slope.
In some embodiments, the memory includes instructions stored thereon, which when executed by the processor, cause the processor to refine raw data from the second segment by extrapolating the association curve fit to the first segment through the second segment and subtracting the extrapolated association curve from the raw data of the second segment, wherein the line is fit to the refined second segment data.
In some embodiments, the memory includes instructions stored thereon, which when executed by the processor, cause the processor to section the interferometry data into three consecutive segments, wherein a one phase exponential decay curve is fit to raw data from the third segment.
In some embodiments, the memory includes instructions stored thereon, which when executed by the processor, cause the processor to determine a goodness of fit metric for each fitted curve. In some embodiments, the goodness of fit metric is an r-squared value. In some embodiments, the memory includes instructions stored thereon, which when executed by the processor, cause the processor to detect negatives of the antibody copies not binding to the target of interest, false positives of non-specific binding proteins binding to the target of interest, and/or hits of the antibody copies binding to the target of interest. In some embodiments, negatives and false positives are detected by determining if the slope of the line fit to the second segment is below a predetermined threshold value and/or if the r-squared value of the line fit to the second segment is below a predetermined threshold value. In some embodiments, hits are detected by determining if the slope of the line fit to the second segment meets of exceeds a predetermined threshold value and if the r-squared value of the line fit to the second segment meets or exceeds a predetermined threshold value. In some embodiments, whether or not the sensor tip is saturated by antibody copies is determined by calculating a slope of the extrapolated association curve and determining if the slope of the extrapolated association curve meets or exceeds a predetermined threshold value.
In some embodiments, the system is configured to identify antibodies with affinity to the target of interest for a plurality of inputs each including sequential wavelength shift measurements resulting from interactions between a sensor tip including a target of interest, copies of an antibody, and tag binding antibodies. In some embodiments, the copies for each input are of a different antibody. In some embodiments, the inputs are ranked based on a binding kinetic parameter predicted for each of the different antibodies to the target of interest. In some embodiments, the binding kinetic parameter is a dissociation rate constant.
In some embodiments, the system further includes a bio-layer interferometry sensor tip configured to generate the raw bio-layer interferometry data and transmit it to the processor, the sensor tip including: a surface including the target of interest; a light source configured to irradiate the surface; and a detector configured to detect an interaction between antibodies and the target of interest.
In some embodiments, the system further includes a sorting flow cytometer including: a light source configured to irradiate particles in a flow stream; a detector configured to detect fluorescence of a particle in the flow stream; a droplet stream generator; and an electrostatic droplet deflector for producing a flow cytometrically sorted sample. The sorting flow cytometer may include any one of the sorting flow cytometer as discussed above. In some embodiments, the system may further include a device for performing the BLI screening methods, the device including: a sensor tip including: a surface configured to adhere a target of interest; a light source configured to irradiate the surface; and a detector configured to detect light reflected from the surface. The surface configured to adhere a target of interest may adhere multiple copies of the target of interest such as, e.g., two or more copies, or five or more, or twenty or more, or one hundred or more, or five hundred or more. The device may further include: a first well configured to hold a first aliquot of a solution including copies of a first antibody; a second well configured to hold a second aliquot if the solution including copies of the first antibody and a second antibody configured to bind to the first antibody; a third well configured to hold a buffer including a lower concentration of the first antibody copies than the second aliquot of the solution; and a means for transferring the sensor tip to the first well, transferring the sensor tip from the first well to the second well, and transferring the sensor tip from the second well to the third well.
In some embodiments, the means for transferring may be a robotic arm configured to move the sensor tip from well to well. In some embodiments, the means may be a track configured to move each well to the sensor tip. In some instances, the volumes of liquid may be flowed to the sensor tip and tag binding antibodies may be injected into the first well after a predetermined amount of time.
In some instances, the systems further include one or more computers for complete automation or partial automation of the methods described herein. In some embodiments, systems include a computer having a computer readable storage medium with a computer program stored thereon.
In embodiments, the system includes an input module, a processing module and an output module. The subject systems may include both hardware and software components, where the hardware components may take the form of one or more platforms, e.g., in the form of servers, such that the functional elements, i.e., those elements of the system that carry out specific tasks (such as managing input and output of information, processing information, etc.) of the system may be carried out by the execution of software applications on and across the one or more computer platforms represented of the system.
Systems may include a display and operator input device. Operator input devices may, for example, be a touchscreen, a keyboard, a mouse, or the like. The processing module includes a processor which has access to a memory having instructions stored thereon for performing the steps of the subject methods. The processing module may include an operating system, a graphical user interface (GUI) controller, a system memory, memory storage devices, and inputoutput controllers, cache memory, a data backup unit, and many other devices. The processor may be a commercially available processor or it may be one of other processors that are or will become available. The processor executes the operating system and the operating system interfaces with firmware and hardware in a well-known manner, and facilitates the processor in coordinating and executing the functions of various computer programs that may be written in a variety of programming languages, such as Java, Perl, C, C++, Python, other high-level or low- level languages, as well as combinations thereof, as is known in the art. The operating system, typically in cooperation with the processor, coordinates and executes functions of the other components of the computer. The operating system also provides scheduling, input-output control, file and data management, memory management, and communication control and related services, all in accordance with known techniques. The processor may be any suitable analog or digital system. In some embodiments, the processor includes analog electronics which provide feedback control, such as for example positive or negative feedback control.
The system memory may be any one of a variety of known or future memory storage devices. Examples include any commonly available random access memory (RAM), magnetic medium such as a resident hard disk or tape, an optical medium such as a read and write compact disc, flash memory devices, or other memory storage device. The memory storage device may be any one of a variety of known or future devices, including a compact disk drive, a tape drive, a removable hard disk drive, or a diskette drive. Such types of memory storage devices typically read from, and/or write to, a program storage medium (not shown) such as, respectively, a compact disk, magnetic tape, removable hard disk, or floppy diskette. Any one of these program storage media, or others now in use or that may later be developed, may be considered a computer program product. As will be appreciated, these program storage media typically store a computer software program and/or data. Computer software programs, also called computer control logic, typically are stored in system memory and/or the program storage device used in conjunction with the memory storage device.
In some embodiments, a computer program product is described including a computer usable medium having control logic (computer software program, including program code) stored therein. The control logic, when executed by the processor the computer, causes the processor to perform functions described herein. In other embodiments, some functions arc implemented primarily in hardware using, for example, a hardware state machine.
Implementation of the hardware state machine so as to perform the functions described herein will be apparent to those skilled in the relevant arts.
Memory may be any suitable device in which the processor can store and retrieve data, such as magnetic, optical, or solid-state storage devices (including magnetic or optical disks or tape or RAM, or any other suitable device, either fixed or portable). The processor may include a general-purpose digital microprocessor suitably programmed from a computer readable medium carrying necessary program code. Programming can be provided remotely to processor through a communication channel, or previously saved in a computer program product such as memory or some other portable or fixed computer readable storage medium using any one of those devices in connection with memory. For example, a magnetic or optical disk may carry the programming, and can be read by a disk writer/reader. Systems of the invention also include programming, e.g., in the form of computer program products, algorithms for use in practicing the methods as described above. Programming according to the present invention can be recorded on computer readable media, e.g., any medium that can be read and accessed directly by a computer. Such media include, but are not limited to: magnetic storage media, such as floppy discs, hard disc storage medium, and magnetic tape; optical storage media such as CD- ROM; electrical storage media such as RAM and ROM; portable flash drive; and hybrids of these categories such as magnetic/optical storage media.
The processor may also have access to a communication channel to communicate with a user at a remote location. By remote location is meant the user is not directly in contact with the system and relays input information to an input manager from an external device, such as a computer connected to a Wide Area Network (“WAN”), telephone network, satellite network, or any other suitable communication channel, including a mobile telephone (i.e., smartphone).
In some embodiments, systems according to the present disclosure may be configured to include a communication interface. In some embodiments, the communication interface includes a receiver and/or transmitter for communicating with a network and/or another device. The communication interface can be configured for wired or wireless communication, including, but not limited to, radio frequency (RF) communication (e.g., Radio-Frequency Identification (RFID), Zigbee communication protocols, Z-Wave communication protocols, ANT communication protocols, WiFi, infrared, wireless Universal Serial Bus (USB), Ultra Wide Band (UWB), Bluetooth® communication protocols, and cellular communication, such as code division multiple access (CDMA) or Global System for Mobile communications (GSM).
In one embodiment, the communication interface is configured to include one or more communication ports, e.g., physical ports or interfaces such as a USB port, an RS-232 port, or any other suitable electrical connection port to allow data communication between the subject systems and other external devices such as a computer terminal (for example, at a physician’s office or in hospital environment) that is configured for similar complementary data communication.
In one embodiment, the communication interface is configured for infrared communication, Bluetooth® communication, or any other suitable wireless communication protocol to enable the subject systems to communicate with other devices such as computer terminals and/or networks, communication enabled mobile telephones, personal digital assistants, or any other communication devices which the user may use in conjunction.
In one embodiment, the communication interface is configured to provide a connection for data transfer utilizing Internet Protocol (IP) through a cell phone network, Short Message Service (SMS), wireless connection to a personal computer (PC) on a Local Area Network (LAN) which is connected to the internet, or WiFi connection to the internet at a WiFi hotspot.
In one embodiment, the subject systems are configured to wirelessly communicate with a server device via the communication interface, e.g., using a common standard such as 802.11 or Bluetooth® RF protocol, or an IrDA infrared protocol. The server device may be another portable device, such as a smart phone, Personal Digital Assistant (PDA) or notebook computer; or a larger device such as a desktop computer, appliance, etc. In some embodiments, the server device has a display, such as a liquid crystal display (LCD), as well as an input device, such as buttons, a keyboard, mouse or touch-screen.
In some embodiments, the communication interface is configured to automatically or semi-automatically communicate data stored in the subject systems, e.g., in an optional data storage unit, with a network or server device using one or more of the communication protocols and/or mechanisms described above. Output controllers may include controllers for any one of a variety of known display devices for presenting information to a user, whether a human or a machine, whether local or remote. If one of the display devices provides visual information, this information typically may be logically and/or physically organized as an array of picture elements. A graphical user interface (GUI) controller may include any one of a variety of known or future software programs for providing graphical input and output interfaces between the system and a user, and for processing user inputs. The functional elements of the computer may communicate with each other via system bus. Some of these communications may be accomplished in alternative embodiments using network or other types of remote communications. The output manager may also provide information generated by the processing module to a user at a remote location, e.g., over the Internet, phone or satellite network, in accordance with known techniques. The presentation of data by the output manager may be implemented in accordance with a variety of known techniques. As some examples, data may include CSV, SQL, HTML or XML documents, email or other files, or data in other forms. The data may include Internet URL addresses so that a user may retrieve additional CSV, SQL, HTML, XML, or other documents or data from remote sources. The one or more platforms present in the subject systems may be any type of known computer platform or a type to be developed in the future, although they typically will be of a class of computer commonly referred to as servers. However, they may also be a main-frame computer, a workstation, or other computer type. They may be connected via any known or future type of cabling or other communication system including wireless systems, either networked or otherwise. They may be co-located or they may be physically separated. Various operating systems may be employed on any one of the computer platforms, possibly depending on the type and/or make of computer platform chosen. Appropriate operating systems include Windows, iOS, macOS, watchOS, Android, Oracle Solaris, Linux, IBM i, Unix, and others.
Aspects of the present disclosure further include non-transitory computer readable storage mediums having instructions for practicing the subject methods. Computer readable storage mediums may be employed on one or more computers for complete automation or partial automation of a system for practicing methods described herein. In certain embodiments, instructions in accordance with the method described herein can be coded onto a computer- readable medium in the form of “programming”, where the term "computer readable medium" as used herein refers to any non-transitory storage medium that participates in providing instructions and data to a computer for execution and processing. Examples of suitable non- transitory storage media include a floppy disk, hard disk, optical disk, magneto-optical disk, CD- ROM, CD-R, magnetic tape, non-volatile memory card, ROM, DVD-ROM, Blue-ray disk, solid state disk, and network attached storage (NAS), whether or not such devices are internal or external to the computer. A file containing information can be “stored” on computer readable medium, where “storing” means recording information such that it is accessible and retrievable at a later date by a computer. The computer-implemented method described herein can be executed using programming that can be written in one or more of any number of computer programming languages. Such languages include, for example, Python, Java, Java Script, C, C#, C++, Go, R, Swift, PHP, as well as many others.
The non-transitory computer readable storage medium may be employed on one or more computer systems having a display and operator input device. Operator input devices may, for example, be a keyboard, mouse, or the like. The processing module includes a processor which has access to a memory having instructions stored thereon for performing the steps of the subject methods. The processing module may include an operating system, a graphical user interface (GUI) controller, a system memory, memory storage devices, and input-output controllers, cache memory, a data backup unit, and many other devices. The processor may be a commercially available processor or it may be one of other processors that are or will become available. The processor executes the operating system and the operating system interfaces with firmware and hardware in a well-known manner, and facilitates the processor in coordinating and executing the functions of various computer programs that may be written in a variety of programming languages, such as those mentioned above, other high level or low level languages, as well as combinations thereof, as is known in the art. The operating system, typically in cooperation with the processor, coordinates and executes functions of the other components of the computer. The operating system also provides scheduling, input-output control, file and data management, memory management, and communication control and related services, all in accordance with known techniques.
UTILITY
The methods and systems of the invention, e.g., as described above, find use in a variety of applications where it is desirable to identify rare high-affinity antibodies against challenging targets. Embodiments of the present disclosure find use in applications wherein it is desired to identify antibodies against antigens previously beyond the scope of immunization. In some embodiments, the subject methods and systems may facilitate identification of antibodies with lower kinetic off-rates, enhanced screening sensitivity, and a reduction in time to identify antibodies of interest.
EXEMPLARY NON-LIMITING ASPECTS OF THE DISCLOSURE
Aspects, including embodiments, of the present subject matter described above may be beneficial alone or in combination, with one or more other aspects or embodiments. Without limiting the foregoing description, certain non-limiting aspects of the disclosure are provided below. As will be apparent to those of ordinary skill in the art upon reading this disclosure, each of the individually numbered aspects may be used or combined with any one of the preceding or following individually numbered aspects. This is intended to provide support for all such combinations of aspects and is not limited to combinations of aspects explicitly provided below. It will be apparent to one of ordinary skill in the art that various changes and modifications can be made without departing from the spirit or scope of the invention.
1. A method for identifying an antibody with affinity to a target of interest by using biolayer interferometry (BLI), the method comprising: contacting a sensor tip comprising the target of interest immobilized thereon with a first aliquot of a solution comprising multiple copies of a first antibody; irradiating the sensor tip with an incident light and measuring a first reflected light, wherein a wavelength shift of first reflected light compared to the incident light is indicative of binding of the first antibody to the target of interest immobilized on the sensor tip; contacting the sensor tip with a second aliquot of the solution comprising multiple copies of the first antibody, wherein the second aliquot further comprises a second antibody that binds to the first antibody; and irradiating the sensor tip with the incident light and measuring a second reflected light, wherein a wavelength shift of second reflected light compared to the incident light and/or the first reflected light is indicative of binding of the second antibody to the first antibody bound to the sensor tip, wherein presence of: (a) the wavelength shift of first reflected light compared to the incident light and (b) the wavelength shift of the second reflected light compared to the incident light and/or the first reflected light identifies the first antibody as having affinity to the target of interest.
2. The method according to Aspect 1, wherein the method further comprises contacting the sensor tip with a buffer and measuring a dissociation rate of the first antibody from the target of interest.
3. The method according to Aspects 1 or 2, wherein the method is performed simultaneously or sequentially on a plurality of solutions comprising different antibodies.
4. The method according to any one of Aspects 1 to 3, wherein the antibodies are produced from a phage display library.
5. The method according to any one of Aspects 1 to 4, wherein the antibodies are Fab, Fv, scFv, VH domain antibodies, or nanobodies.
6. The method according to any one of Aspects 1-5, wherein the solution comprising multiple copies of the first antibody is generated from a bacteria infected with a phage displaying the first antibody, wherein the phage is identified by a method comprising: fluorescently labeling a phage display library comprising a multitude of different antibodies displayed on phages; contacting the labeled phage display library with the target of interest immobilized on beads to produce bead-phage complexes; and analyzing the bead-phage complexes using flow cytometry to generate a profile of the complexes; using the profile to identify high-affinity antibodies. 7. The method according to Aspect 6, wherein the profile comprises a histogram representing the frequency of complexes having different fluorescence intensities.
8. The method according to Aspect 7, wherein a median fluorescence intensity (MFI) and/or a standard deviation (SD) are calculated for one or more components of the profile.
9. The method according to Aspect 8, wherein the high affinity antibodies correspond to a component of the profile having a high MFI.
10. The method according to Aspects 8 or 9, wherein the high affinity antibodies correspond to a component of the profile having a high SD.
11. The method of according to any one of Aspects 6 to 10, further comprising: sorting the bead-phage complexes based on a level of fluorescent phages bound to the beads, thereby separating complexes exhibiting a threshold level of fluorescence from complexes exhibiting fluorescence below the threshold level.
12. The method according to any one of Aspects 6 to 11, wherein the phage display library comprises phages from two or more rounds of preselection for phages that bind to the target of interest.
13. The method according to Aspect 12, wherein the phage display library comprises phages from two or more rounds of preselection for phages that bind to the target of interest.
14. The method according to Aspects 12 or 13, wherein phages from each round are separately contacted with the target of interest immobilized on beads and analyzed using flow cytometry to generate a profile for phages from each round of preselection.
15. The method according to Aspect 14, comprising using the generated profiles to identify high-affinity antibodies. 16. The method according to Aspect 15, wherein the high-affinity antibodies have the profile with the highest MFI.
17. The method according to any one of Aspects 6 to 16, wherein the phage display library is labeled with fluorescein isothiocyanate (FITC) or an Alexa Fluor (AF) dye.
18. The method according to Aspect 17, wherein the phage display library is labeled with FITC.
19. The method according to Aspect 17, wherein the phage display library is labeled with AF647.
20. The method according to any one of Aspects 6 to 19, wherein the phages are M13 phages.
21. The method according to Aspect 20, wherein the M13 pages are labeled via N-termini and/or lysine residues of the pVIII coat proteins.
22. The method according to any one of Aspects 6 to 21, wherein the phage display library is generated from bacteria infected with a phage.
23. The method according to Aspect 22, wherein the bacteria are Escherichia coli.
24. The method according to Aspects 22 or 23, wherein the solution comprising multiple copies of a first antibody is crude periplasmic extract (PPE) from bacteria infected with a phage.
25. The method according to Aspect 24, wherein the first antibody comprises a tag and the second antibody binds to the tag.
26. The method according to any one of Aspects 2-25, wherein the buffer comprises no detectable copies of the first antibody or the second antibody. 27. The method according to any one of Aspects 2-26, wherein the method further comprises determining a binding kinetic parameter of first antibody to the target of interest using tip measurements.
28. The method according to Aspect 27, wherein the binding kinetic parameter is a dissociation rate constant.
29. The method according to Aspect 28, wherein the dissociation rate constant is determined using measurements generated while the tip is contacting the buffer.
30. The method according to any one of the preceding Aspects, wherein the method further comprises determining false positives using tip measurements.
31. The method according to Aspect 30, wherein the false positives are determined using measurements generated while the tip is contacting the second aliquot of the solution comprising multiple copies of the first antibody and wherein the second aliquot further comprises the second antibody that binds to the first antibody, wherein presence of (a) and absence of (b) identifies the first antibody as a false positive antibody.
32. The method according to any one of the preceding Aspects, wherein the method further comprises determining if the sensor tip is saturated by antibody copies using tip measurements.
33. The method according to Aspect 32, wherein the saturation is determined using measurements generated while the tip is in the first volume of liquid.
34. A system configured to perform the method according to any one of Aspects 1 to 33, optionally wherein the system comprises a biolayer interferometry device and a cell sorter.
35. A biolayer interferometry device configured to perform the screening according to any one of Aspects 1 to 33. 36. A method for identifying antibodies with affinity to a target of interest, the method comprising: conducting a first round of biopanning comprising isolating phages displaying antibodies that bind to the target of interest and fluorescently labeling the phages to generate a first labeled population of phages; conducting a second round of biopanning comprising isolating phages displaying antibodies that bind to the target of interest and fluorescently labeling the phages to generate a second labeled population of phages; separately contacting the first and the second labeled population of phages with the target of interest immobilized on beads to produce bead-phage complexes; analyzing the complexes using flow cytometry to generate a first fluorescence profile of the complexes produced from the first labeled population of phages and a second fluorescence profile of second labeled population of phages; and using the first and second profiles to identify antibodies with affinity to the target of interest.
37. The method according to Aspect 36, wherein the profile comprises a histogram representing the frequency of complexes having different fluorescence intensities.
38. The method according to Aspect 37, wherein a median fluorescence intensity (MFI) and/or a standard deviation (SD) are calculated for one or more components of the profiles.
39. The method according to Aspect 38, wherein the antibodies associated with the profile having the highest MFI are identified as high affinity antibodies.
40. The method according to Aspect 38, wherein when the MFIs of each profile are similar, the antibodies associated with the profile with the highest SD are identified as high affinity antibodies. 41 . The method according to any one of Aspects 38 to 40, wherein the antibodies associated a profile with the highest MFI and SD arc identified as high affinity antibodies.
42. The method of according to any one of Aspects 36 to 41, wherein the isolating further comprises: sorting the complexes based on a level of fluorescent phages bound to the beads, thereby separating complexes exhibiting a threshold level of fluorescence from complexes exhibiting fluorescence below the threshold level; wherein the high affinity antibodies are present in the complexes exhibiting the threshold level of fluorescence.
43. The method according to any one of Aspects 36 to 42, wherein the method comprises conducting three or more rounds of biopanning to generate three or more labeled populations of phages.
44. The method according to Aspect 43, wherein the method comprises conducting four or more rounds of biopanning to generate four or more labeled populations of phages.
45. The method according to any one of Aspects 36 to 44, wherein the method further comprises screening the high affinity antibodies individually to identify antibodies with a high affinity to the target of interest using bio-layer interferometry (BLI).
46. The method according to Aspect 45, wherein the screening comprises: infecting bacteria with phages expressing the high affinity antibodies; separating the bacteria into single cells and growing bacterial colonies that each express a single type of antibody; preparing an extract from the colonies to provide a plurality of solutions, each solution comprising multiple copies of single type of antibody; contacting in parallel, a first aliquot of each of the plurality of solutions with a sensor tip comprising the target of interest immobilized thereon; contacting in parallel, a second aliquot of each of the plurality of solutions with the sensor tip, wherein the second aliquot further comprises a second antibody that binds to each of the single type of antibodies; wherein binding between the target of interest, the antibodies expressed by the phages, and the second are determined by irradiating the target of interest with light and measuring wavelength shift over time using the sensor tip; and wherein a difference in the rate of change of wavelength shift over time between when the sensor tip is in contact with first aliquot and when the sensor tip is in contact with the second aliquot is used to identify whether the antibody expressed by the phages has affinity to the target of interest.
47. The method according to Aspect 46, wherein the method further comprises contacting a third buffer with the sensor tip.
48. The method according to Aspects 46 or 47, wherein the phages produce an antibody comprising a tag and the second antibody binds to the tag.
49. The method according to any one of Aspects 46 to 48, wherein the screening is performed for phages produced from at least 10, at least 30, at least 100, at least 300, or more individual bacterial colonies.
50. The method according to any one of Aspects 36 to 49, wherein the phage display library is labeled with fluorescein isothiocyanate (FITC) or an Alexa Fluor (AF) dye.
51. The method according to Aspect 50, wherein the phage display library is labeled with FITC.
52. The method according to Aspect 50, wherein the phage display library is labeled with 53. The method according to any one of Aspects 36 to 52, wherein the phages are Ml 3 phages.
54. The method according to Aspect 53, wherein the M13 pages are labeled via N-termini and/or lysine residues of the pVIII coat proteins.
55. The method according to any one of Aspects 46 to 54, wherein the phage display library is generated using bacteria of the genus Escherichia.
56. The method according to Aspect 55, wherein the bacteria are Escherichia coli bacteria.
57. The method according to any one of Aspects 44 to 56, wherein the solution comprises crude periplasmic extract (PPE) produced from bacteria.
58. The method according to any one of Aspects 47 to 56, wherein the buffer does not include a chaotropic agent.
59. The method according to any one of Aspects 47 to 58, wherein the buffer comprises a chaotropic agent.
60. The method according to any one of Aspects 47 to 59, wherein the method further comprises determining a binding kinetic parameter of the antibodies to the target of interest using tip measurements.
61. The method according to Aspect 60, wherein the binding kinetic parameter is a dissociation rate constant.
62. The method according to Aspect 61, wherein the dissociation rate constant is determined using measurements generated while the tip is in the buffer. 63. The method according to any one of Aspects 46 to 62, wherein the method further comprises determining false positives using tip measurements.
64. The method according to Aspect 63, wherein the false positives are determined using measurements generated while the tip is in the second aliquot.
65. The method according to any one of Aspects 46 to 64, wherein the method further comprises determining if the sensor tip is saturated by antibody copies using tip measurements.
66. The method according to Aspect 65, wherein the saturation is determined using measurements generated while the tip is in the first volume of liquid.
67. A system configured to perform the method according to any one of Aspects 36 to 66.
68. A biolayer interferometry device configured to perform the screening according to any one of Aspects 35 to 66.
69. A system for identifying antibodies with affinity to a target of interest, the system comprising: a processor comprising memory operably coupled to the processor wherein the memory comprises instructions stored thereon, which when executed by the processor, cause the processor to: obtain raw bio-layer interferometry data, wherein the interferometry data comprises sequential wavelength shift measurements resulting from interactions between a sensor tip comprising a target of interest, copies of an antibody, and tag binding antibodies; section the interferometry data into two consecutive segments; fit a one phase exponential association curve to raw data from the first segment, wherein the sequentially later end of the one phase exponential association curve comprises a first slope; fit a line to the second segment data, wherein the fitted line comprises a second slope; and compare the first slope to the second slope, wherein the antibody copies are identified as having affinity to the target of interest if the second slope is greater than the first slope.
70. The system according to Aspect 69, wherein the memory comprises instructions stored thereon, which when executed by the processor, cause the processor to refine raw data from the second segment by extrapolating the association curve fit to the first segment through the second segment and subtracting the extrapolated association curve from the raw data of the second segment, wherein the line is fit to the refined second segment data.
71. The system according to Aspects 69 or 70, wherein the memory comprises instructions stored thereon, which when executed by the processor, cause the processor to section the interferometry data into three consecutive segments, wherein a one phase exponential decay curve is fit to raw data from the third segment.
72. The system according to Aspect 71, wherein the memory comprises instructions stored thereon, which when executed by the processor, cause the processor to determine a goodness of fit metric for each fitted curve.
73. The system according to Aspect 72, wherein the goodness of fit metric is an r-squared value.
74. The system according to Aspect 73, wherein the memory comprises instructions stored thereon, which when executed by the processor, cause the processor to detect negatives of the antibody copies not binding to the target of interest, false positives of non-specific binding proteins binding to the target of interest, and/or hits of the antibody copies binding to the target of interest. 75. The system according to Aspect 74, wherein negatives and false positives are detected by determining if the slope of the line fit to the second segment is below a predetermined threshold value.
76. The system according to Aspect 74 or 75, wherein negatives and false positives are detected by determining if the r- squared value of the line fit to the second segment is below a predetermined threshold value.
77. The system according to any one of Aspect 74 to 76, wherein false positives are detected by determining if the r-squared value of the association curve fit to the first segment meets or exceeds a predetermined threshold value.
78. The system according to any one of Aspect 74 to 76, wherein negatives are detected by determining if the r-squared value of the association curve fit to the first segment is below a predetermined threshold value.
79. The system according to any one of Aspect 74 to 78, wherein the hits are detected by determining if the slope of the line fit to the second segment meets of exceeds a predetermined threshold value and if the r-squared value of the line fit to the second segment meets or exceeds a predetermined threshold value.
80. The system according to Aspect 79, wherein the memory comprises instructions stored thereon, which when executed by the processor, cause the processor to determine if the sensor tip is saturated by antibody copies by calculating a slope of the extrapolated association curve.
81. The system according to Aspect 80, wherein the sensor tip is determined to be saturated if the slope of the extrapolated association curve meets or exceeds a predetermined threshold value.
82. The system according to any one of the Aspects 79 to 81, wherein the memory comprises instructions stored thereon, which when executed by the processor, cause the processor to determine antibody clone expression by calculating the total wavelength shift of the first segment.
83. The system according to Aspect 82, wherein the antibody copies are determined to have low expression if the total wavelength shift of the first segment is below a predetermined threshold value.
84. The system according to any one of the Aspects 79 to 83, wherein the memory comprises instructions stored thereon, which when executed by the processor, cause the processor to predict a dissociation rate for the antibody copies and the target of interest.
85. The system according to Aspect 84, wherein the dissociation rate is predicted using the exponential decay curve fit to the third segment.
86. The system according to Aspects 84 or 85, wherein the memory comprises instructions stored thereon, which when executed by the processor, cause the processor to determine if the predicted dissociation rate is inaccurate.
87. The system according to Aspect 86, wherein the dissociation rate is determined to be inaccurate if the r-squared value of the exponential decay curve fit to the third segment is below a predetermined threshold value.
88. The system according to any one of the Aspects 69 to 87, wherein the system is configured to identify antibodies with affinity to the target of interest for a plurality of inputs each comprising sequential wavelength shift measurements resulting from interactions between a sensor tip comprising a target of interest, copies of an antibody, and tag binding antibodies.
89. The system according to Aspect 88, wherein the copies for each input are of a different antibody. 90. The system according to Aspects 88 or 89, wherein the memory comprises instructions stored thereon, which when executed by the processor, cause the processor to rank the inputs based on a binding kinetic parameter predicted for each of the different antibodies to the target of interest.
91. The system according to Aspect 90, wherein the binding kinetic parameter is a dissociation rate constant.
92. The system according to any one of the Aspects 69 to 91, wherein the system further comprises a bio-layer interferometry sensor tip configured to generate the raw bio-layer interferometry data and transmit it to the processor, the sensor tip comprising: a surface comprising the target of interest; a light source configured to irradiate the surface; and a detector configured to detect an interaction between antibodies and the target of interest.
EXAMPLES
As demonstrated in the above disclosure, the present invention has a wide variety of applications. The following examples are put forth so as to provide those of ordinary skill in the art with a complete disclosure and description of how to make and use the present invention and are not intended to limit the scope of what the inventors regard as their invention nor are they intended to represent that the experiments below are all or the only experiments performed. Those of skill in the art will readily recognize a variety of noncritical parameters that could be changed or modified to yield essentially similar results. Efforts have been made to ensure accuracy with respect to numbers used (e.g. amounts, volumes, etc.) but some experimental errors and deviations should be accounted for.
Introduction
In vitro biopanning platforms using synthetic phage display antibody libraries have enabled the identification of antibodies against antigens that were once thought to be beyond the scope of immunization. Expanding these methods to identifying rare high-affinity binders against challenging targets remains a critical challenge. Here, a new biopanning pipeline, RAPID (Rare Antibody Phage Isolation and Discrimination), is presented for the identification of rare high- affinity antibodies against challenging targets. RAPID biopanning uses fluorescent labeled phage displayed fragment antigen-binding (Fab) antibody libraries for the isolation of high-affinity binders with fluorescent activated sorting. Subsequently, discriminatory hit screening is performed with a newly developed biolayer interferometry (BLI) method, BIAS (Biolayer Interferometry Antibody Screen), where candidate binders are ranked and prioritized according to their estimated kinetic off rates. A well characterized antibody-antigen pair, P1A4 and ARS1620, were used to establish the pipeline, and two challenging targets (CS3D, and CHIP), were employed, where RAPID biopanning enabled for the identification of high-affinity antibodies.
Antibodies (Abs) have and continue to be one of the most invaluable tools for biological research1. In particular, the utility of rapidly generated high-quality Abs has been highlighted in the 2020 SARS-CoV-2 pandemic as attractive therapeutic interventions23,4. While immunization has been the traditional method for Ab discovery, biopanning using synthetically displayed Ab libraries (i.e., phage display and yeast surface display) have expanded the field and allowed for non-protcin antigen (Ag) targets such as DNA5 and RNA6 to be targeted.
Standard phage display biopanning methods largely consist of two stages: (1) Iterative rounds of washing and amplification of displayed Abs bound to immobilized Ag to achieve an enriched population of higher- affinity binders, and (2) Random screening of enriched pools of displayed Abs for the identification of high-affinity Ab clones (hits). Notably, the in vitro selection and screening nature of the method enables the identification of high-affinity Abs in conditions otherwise impossible with immunization (i.e., changes in pH, temperature, oligomeric complex states, ligand states etc.),
Despite these attractive features of biopanning, it remains challenging to identify, and characterize selective, high-affinity Abs where their prevalence’s are extremely low (i.e., target Ag exhibits low antigenicity, and/or conformational heterogeneity). Poor enrichment towards Ag-Ab binding in turn allows additional factors (i.e., amplification discrepancies of phagemid- containing E. coli and inconsistent display propensities of phage) to undermine the enrichment process resulting in an overall depletion of promising binders with continued rounds of selection. In turn, significantly larger number of clones need to be screened and prioritized, and as this process largely relies on stochastic clone picking, the vast majority of candidate clones, and potentially rare high-affinity binders can be left unexamined.
These challenges have been met with some effective solutions such as improvements in synthetic Ab library design7, selective isolation of high-affinity binders using fluorescence- activated cell sorting (FACS)8 coupled with yeast surface display (YSD9 10), and selection-based methods for identifying candidate clones11. Yet, further improvements in the current platforms for efficient recombinant Ab discovery are crucial for continued expansion of the field.
Here a novel phage display pipeline, RAPID (Rare Antibody Phage Isolation and Discrimination), is provided for identifying rare high-affinity Abs against challenging targets using a recombinant phage displayed Fab (ph-Fab) library (Fig. 1). The RAPID biopanning pipeline was strategized to selectively isolate the most promising subset population of binders and subsequently perform screening in a discriminatory fashion allowing for a highly efficient strategy for the identification of high- affinity Fabs. This is distinct from the standard biopanning pipeline where the total population of enriched displayed Abs are non-discriminatorily screened. RAPID biopanning utilizes fluorescent labeling of ph-Fab libraries to precisely identify and isolate populations of rare high-affinity ph-Fabs, and a newly developed BLI (Biolayer interferometry) method called BIAS (Biolaycr Interferometry Antibody Screen) is used for discriminatory screening of candidate clones. (Fig. 1).
RAPID biopanning combines the advantages of phage display by utilizing large library sizes (up to IO10) while leveraging the quantitative binding screening. In addition, RAPID is not material intensive (~ 20 ug of Ag required) and therefore can be used for targets that are difficult to acquire reagent quantities of. Overall, the RAPID pipeline is widely applicable and allows for a highly efficient strategy for the identification of rare high-affinity binders.
Here, controls for each step of RAPID is first provided using a well characterized antibody, P1A4, which binds ARS1620, a preclinical, covalent KRas G12C inhibitor. Subsequently, two challenging biological panning campaigns are described as examples to highlight the power of RAPID biopanning. Cyclic STAT3 Decoy (CS3D) is a novel double stranded DNA drug candidate, and there is limited precedence for the description of Fabs that bind to dsDNA antigens. Carboxyl terminus of Hsp-70 interacting protein (CHIP) is a E3 ubiquitin ligase target that is highly conformationally diverse and for which previous in-house standard panning protocols failed to identify binders against.
Example 1: The Rare Antibody Phage Isolation and Discrimination (RAPID) pipeline
The RAPID biopanning pipeline comprises of four steps: (1) Fluorescent labeling of ph- Fab libraries for the detection and quantification of ph-Fab-Ag-bead complexes (2) Biopanning campaign enrichment progression profiling with flow cytometry for the accurate determination of Ag-Fab enrichment, (3) Isolation of high-affinity binders with fluorescence activated sorting, and (4) Discriminatory hit screening of candidate clones with BIAS (Fig. 1).
NHS-FITC labeling ofph-Fab enables quantitative detection ofFab-Ag binding
For the RAPID pipeline to be a feasible protocol, fluorescence from Ag-Bead-phage complexes need to directly correlate to the number of ph-Fabs bound per Ag-Bead. In addition, labeling should occur with minimal variability between different ph-Fabs and not disrupt the interactions between displayed Fabs and bead immobilized Ags, so as to not introduce additional biases. To ensure that fluorescent intensities correlate to the ph-Fab affinity, the method was optimized with four unique ph-Fabs (ph-PlH6, ph-PICl, ph-P2Fl 1, and phP2B2) that bind to a common Ag, an ARS1620-labeled KRas G12C peptide VVVGACGVGK (SEQ ID NO:1) (ARS 1620-V7) (Fig. 14). The duration of ph-Fab incubation with Ag-beads was examined to avoid the over- saturation of Ag-bcads with bound ph-Fabs.
Using two FITC-labeled ph-Fabs with varying affinities (ph-P2B2 and ph-PlA4), an incubation period of 1 hour generated a fluorescent signal that was sufficiently high. An additional 2 hours of incubation resulted in an increase in fluorescent intensity, indicating that the Ag-beads were not saturated after the initial 1-hour incubation (Fig. 14a).
The density of Ag molecules immobilized on beads was also considered to achieve a 1 : 1 binding mode to ph-Fab, which would ensure that the fluorescent signal observed on each bead accurately reflected the potential maximal quantitative number of ph-Fabs bound. From immobilizing biotin-FITC to streptavidin-beads, biotin-FITC immobilization saturated near a final concentration of 0.1 pM, and less than half of the maximum amount was shown to immobilize at 0.01 pM according to the normalized MFI (Fig. 8). An antigen concentration of 0.01 pM was used for all subsequent RAPID flow cytometry and FACS experiments. Results indicate that no self-quenching occurs from proximally immobilized FITC molecules at the upper limits of fluorescent signal.
Applying these conditions, the four different ph-Fabs were FITC labeled separately and incubated individually with Ag-Beads (Fig. 14b). Good agreement was observed between the fluorescent intensity and the affinity of the displayed Fab where a ~3-fold difference in MFI is observed between the lowest-affinity binder (ph-P2B2, Kd = 186.8 nM) and the highest-affinity binder (ph-P2Fl 1, Kd = 22.5 nM). Furthermore, when examining ph-Fabs with similar affinities (ph-PlH6, Kd = 41.5 nM, ph-PICl Kd = 32.5 nM, ph-P2Fl l Kd = 22.5 nM), consistent patterns emerge between the displayed Kd values and the MFI values. This agreement indicates that NHS labeling of ph-Fabs is indeed a reliable and robust technique for fluorescently labeling M13 phage, facilitating their detection in flow cytometry and FACS.
Additionally, a set of four ph-Fabs consisting of ph-P!A4, ph-PICl, ph-PlH6, and ph- P2F11, each of which binds to the same Ag (ARS1620) were chosen to test if FITC labeling showed any significant variability between different ph-Fabs or if labeling affected ph-Fab binding to bead immobilized Ag. Pull down experiments of different ph-Fabs against the same Ag (ARS1620 peptide) show similar titers of bound phage between labeled and unlabeled phage, indicating that FITC labeling does not significantly disrupt displayed Fab-Ag binding (Fig 2a). Comparing specific labeling parameters, ph-PlA4 and ph-P2Fl l showed variations in both degrees of labeling (60-100 FITC per phage) and normalized fluorescence (-3000-4500 RFU) (Fig 2b). However, given the pull-down titer ratio of labeled ph-PlA4 versus labeled ph- P2Fl l(2.05-fold) roughly match that of the FITC median fluorescence intensity (MFI) ratio between the two ph-Fabs (2.64-fold), these variations can be considered inconsequential (Fig 2a, 2c, 2d).
RAPID flow cytometry for profiling the enrichment of a biopanning campaign.
Having established that the MFI correlates to the binding affinity of ph-Fabs, these flow cytometry methods were applied for the characterization of heterogenous ph-Fab populations meant to model those isolated during a biopanning campaign. Mixtures of ph-Fab containing varying ratios of Ml 3 phage and ph-Pl A4 were employed to emulate progressive rounds of biopanning, wherein a high-affinity binder (ph-PlA4) is gradually enriched within a pool predominantly comprised of nonbinders (Ml 3 phage). These mixtures were then individually FITC labeled, and flow cytometry analysis was performed with ARS1620 bound beads. A normalized MFI of 1.26 is exhibited with 10% ph-Pl A4 when compared to the 0% ph-Pl A4 sample (Figs. 2e, 2f and 14c). Normalized MFI values for 50% and 100% were 2.38 and 3.65 respectively, confirming that enriched populations have higher fluorescent signal. The range of normalized MFI values act as standards in determining future approximate hit rates of a library of ph-Fab with similar kinetic properties as ph-Pl A4 (Kd - 24 nM).
Similarly, by FITC labeling ph-Fab libraries from biopanning campaigns and analyzing by flow cytometry, an accurate, and quantitative distribution of ph-Fab bound to Ag-beads can be observed which is not achievable by previous techniques (i.e., phage ELISA). Applying this to successive rounds of biopanning in a single campaign allows for the distribution of binding to be monitored over time (Fig. 9), allowing the identification of biopanning rounds enriched for binders. The normalized median fluorescent intensity (NMFI) indicates the average affinity of the bound ph-Fab population. Additionally, assuming the NMFI of two different populations are similar, the standard deviation (SD) represents the range of affinities of individual binders. By combining this information, dominant factors that influenced the enrichment process during the biopanning campaign can be determined (Fig. 9). From biopanning profiling data, the round which is the most enriched for Ag-Ab binding can be directly identified and prioritized for screening with BIAS. RAPID FACS allows for the isolation of ph-Fab populations highly enriched for Ag binding.
For cases where significant global Ag-Ab enrichment is not observed, fluorescent bead populations can be sorted using FACS (Fig. 1). A key feature of the RAPID pipeline is the isolation of Ag-Beads exhibiting higher fluorescent signals which harbor bound ph-Fab populations that are enriched with high affinity binders, which would otherwise exist at low frequencies within the initial binder pool.
To confirm this, a mixture of a high and low-affinity ph-Fab binders was generated to represent ph-Fab libraries containing rare high-affinity binders in low frequencies (Fig. 14d). The fluorescent distribution of Ag-Beads agrees with the binding affinity of the bound ph-Fabs. For ph-P2B2 (Kd =186.8 nM, MFI =1147, SD =295), the distribution demonstrates an MFI with a low SD, indicating a more uniform distribution of ph-Fabs. Conversely, with ph-PlH6 (Kd =41.5 nM, MFI =3901, SD =4117), the distribution displays a higher MFI and a higher SD, suggesting a less even distribution of ph-Fabs. This observation can be rationalized by considering that weaker binders have a greater capacity to redistribute their populations compared to stronger binders in a pre-equilibrium system. This is consistent with the results observed when a mixture of ph-P2B2 (90%) and ph-PlH6 (10%) is examined as the distribution exhibits an intermediate MFI and SD, relative to the pure samples (MFI =1446, SD =552).
Furthermore, the correlation between higher fluorescent signals and enriched populations of high-affinity ph-Fabs was investigated by generating a mixture of ph-Fab comprising of a majority population (90%) and a minority population (10%) that were labeled separately with FITC and AF647 respectively, and thereafter combined (Fig. 14e). A low or high affinity binder (ph-P2B2, Kd = 186.8 nM, ph-PlH6, Kd = 41.5 nM) was used as the minority population, while the majority population was kept constant as the low-affinity ph-P2B2. In parallel, both mixtures were incubated with Ag-beads and FITC (majority population) and AF647 (minority population) fluorescent distributions were analyzed by flow cytometry.
The distribution of the majority population is near identical between the two mixtures according to their FITC-MFI (ph-PlH6 mixture MFI =1118, ph-P2B2 mixture MFI =1095), while an increase in fluorescent intensity and SD is observed for the ph-PlH6 containing mixture, shown by the increase in AF647-MIF and AF647-SD (ph-PlH6 mixture MFI =3327, ph-P2B2 mixture MFI =987), (ph-PlH6 mixture SD =2338, ph-P2B2 mixture SD =932). This indicates that the binding events of low or high affinity binders occur independently and retain their respective binding distribution characteristics. Specifically, Ag-Beads are evenly occupied by the low-affinity binder, while simultaneously high-affinity binders arc unevenly distributed. As a result, beads with higher fluorescent signals demonstrate increased intensities, primarily due to the greater abundance of higher affinity binders, rather than the redistribution of the lower affinity binders. The results confirm that Ag-Beads exhibiting higher fluorescent signals are indeed associated with greater enriched populations of higher-affinity ph-Fabs, and isolation of these populations can be achieved with FACS. In this way, it is possible to perform iterative rounds of ph-Fab labeling followed by fluorescence activated sorting, to rescue the enrichment process from potentially severe growth/display propensities and shift the selective pressure back to Ab-Ag binding. On some occasions, minor higher, or shouldering fluorescent populations containing greater numbers of rare high-affinity binders can be sorted (Figs. 1 and 3).
Example 2: Biolayer interferometry antibody screen (BIAS) for discriminatory screening of candidate binders
Despite the capacity of both RAPID flow cytometry and FACS to facilitate the identification and isolation of considerably enriched populations of ph-Fab bound to Ag-Beads, it does not guarantee the exclusion of weaker binders from the isolated pool. To address this, BIAS was developed for the discriminatory screening of candidate Abs by analyzing the real time binding of individual candidate Abs directly from crude periplasmic extract (PPE) samples using BLI. This is distinct from commonly utilized methods for candidate binder screening (i.e., dot blot, ELISA, DNA sequencing) where hits are not distinguishable based on their binding properties. Additionally, BIAS only requires a maximum of ~3 mg of Ag to assay 95 candidate clones which can be screened within 3 hours, making this technique amenable to targets that are difficult to express/purify. BIAS can also be applied to a variety of different assay conditions (i.e., temperature, buffer etc.) and in addition to binding properties of candidate clones, candidate binders that exhibit severely low expression levels can also be identified for deprioritization.
BIAS consists of three steps (Fig 4a). (i) Association- 1 (Assoc- 1): Ag loaded tips are transferred to crude PPEs containing expressed Fabs allowing Fabs to associate to the Ag, (ii) Association-2 (Assoc-2): tips are transferred to wells containing identical PPE + anti-tag IgG, where the anti-tag IgG can bind to the Fab (iii) Dissociation (Dissoc): tips (ii) are Iran si erred to wells containing buffer where Fabs are dissociated. BIAS is able to identify true positive binders from a characteristic association curve observed in the Assoc-2 step which is absent for false positive clones.
In the case that an observed association in Assoc- 1 is occurring from a Fab, the secondary IgG will bind to its tag. A false positive signal in Assoc- 1 arising from non-specific binding of periplasmic proteins will not show this second association signal as these non-specific proteins lack this tag. For these purposes, the entire ph-Fab library includes a myc-tag, therefore, the anti- myc IgG, 9E10, was employed for the Assoc-2 step. This Ab has been reported to be highly specific where one amino acid change has shown to disrupt binding14 therefore binding in Assoc- 2 is a direct readout of tagged Fab binding.
An in-house developed script, BIAS Algorithm Triaging Confirmed Hits (BATCH), individually analyzes the binding curves and categorizes each individual clone into “hits”, “false positives”, and “negatives” (Fig. 10). For “hits”, predicted koffS were used as the distinguishing factor for rank ordering binders as kou predictions are inaccurate due to the presence of inconsistent PPE in the Assoc-1 step (FIG. 11).
Characterizing BIAS with P1A4 and ARS1620 labeled peptide.
To establish proof of concept of the BIAS assay, the Fab-Ag pair, P1A4-ARS1620, was used (koff = 2.07 x 10'3 1/s, Ka= 25.1 nM). P1A4 was tested against an ARS1620 labeled peptide at 250 nM (both Assoc-1 and Assoc-2) where the anti-myc IgG (9E10) was either included or absent in Assoc-2. Solutions of P1A4 spiked into both PBS and crude PPE derived from TGI cells (E. coll) were tested in parallel to determine whether the method could tolerate the presence of the highly heterogenous PPE. An association curve is clearly observed in the Assoc-2 phase in samples that contains the secondary IgG, 9E10, when compared to the samples that do not (Fig 4b, 4c) confirming that true positives are identifiable in the Assoc-2 step. Predicted values of kinetic parameters including koffS from the BATCH yields near identical values to reported koffS tested in vitro indicating that the addition of the Assoc-2 step or the PPE environment does not hinder kinetic predictions (Table 1). Table 1 : BIAS with pure Pl A4 spiked in PBS and PPE.
Figure imgf000065_0001
Table 2. BIAS kinetic parameters (koff, true Assoc-2 slope, and extrapolated assoc-2 linear slope) of multiple concentrations of P1A4 spiked in PPE.
Figure imgf000065_0002
Varying concentrations of P1A4(1O-3OO nM) were also tested to establish concentration dependencies of the assay (Fig. 4d). A series of concentrations of P1A4 spiked in PPE shows an increase in true Assoc-2 slopes as concentration approaches Kd (table 2). Where the Assoc- 1 step is not fully saturated, and the P1A4 sample concentrations of are at non-steady-state conditions, koff predictions are less accurate. To address this, BATCH was developed to flag hits that show low expression or do not fully saturate in Assoc- 1 to indicated inaccuracies in predicted kOft values for the user’s discretion.
RAPID biopanning enables the identification o f rare antibodies against CHIP.
Carboxyl terminus of Hsp-70 interacting protein (CHIP) is an E3 ubiquitin ligase canonically known to interact with heat shock protein 70 (Hsp70) and heat shock protein 90 (Hsp90), leading to the ubiquitination of misfolded clients as well as regulation of chaperone turnover. Recent work demonstrated that CHIP has substrate specificity extending beyond Hsp70/90 and predicted interactions suggest CHIP may have additional Hsp-independent roles in proteostasis and disease states15. To enable further biological studies, Abs against CHIP were developed beyond the available substrate binding inhibitorsl l6. CHIP is a challenging biopanning target due to its homodimeric structure and conformational flexibility, and therefore RAPID biopanning was employed.
Four rounds of standard biopanning were employed against CHIP with increased stringency each round. Subsequently, RAPID flow cytometry was performed by individually FITC labeling each output round. Flow cytometry data shows an increase in normalized MFI from round 2 through 3, and a significant drop is observed at round 4 (Fig 5a). The most enriched round (round 3) shows a normalized MFI of 2.11 which correlates to a reasonably high hit rate between 10 and 50 percent of a high affinity binder (~Kd =25 nM) according to previous control experiments with P1A4 (Fig 14c). As flow cytometry data suggests strong Fab-Ag binding enrichment had occurred, fluorescent activated sorting was deemed unnecessary, and BIAS was performed directly.
95 individual colonies were picked from round 3, and from PPE samples, 18 hits were identified by BIAS, and BATCH calculated kOffS showing a wide range of values (9.40xl06- 5.44xl02 s’1) (Fig. 5b, Fig. 6, Table 3). Candidate hit Fabs that were recombinantly expressed and biochemically characterized exhibited similar trends of koffS compared to those predicted by BIAS (R2 =0.95), and their values were within ~2 fold of each other (Fig 5c). In parallel dot blots and ELIS As were performed to compare BIAS with standard screening methods. Dot blot experiments resulted in 76 hits (80% hit rate) in comparison to 18 hits identified from BIAS (Fig 5d). After exhaustive optimizations, no hits were identified using ELISAs. The biopanning campaign of CHIP demonstrates the power of RAPID biopanning as a method for accurately identifying the most enriched round, determining dominant factors of enrichment, and predicting hit rates of specific rounds of candidate hit screening. Notably, in this example, BIAS is shown to be a critical step in identifying true functional candidate hits and prioritizing potential binders for detailed biochemical characterizations.
Table 3. BIAS hit summary table for CHIP. Inaccurate koff measurements are flagged (*- low expression #-unsaturuated Assoc- 1, !-poor exponential fit for Dissoc). Kd min and max are estimated based on general kon of previously discovered Craik lab Fabs (5 x 104 - 3 x 105)
Figure imgf000066_0001
Figure imgf000067_0001
RAPID identifies rare binders against Gag.
Gaq is a ubiquitous heterotrimeric G protein subunit important in many G protein- coupled receptor signaling cascades. Like other G proteins, the conformation of Gaq is regulated by a guanine nucleotide. While Gaq is normally activated in diverse physiological states, several mutations lead to inappropriate activation of Gaq, leading to challenging diseases like uveal melanoma25 or Sturge Weber syndrome26. To understand how mutations in Gaq lead to activation, it was sought to develop antibody fragments that would enable structural studies. Gaq is a challenging target due to its known dynamic conformational flexibility. Six rounds of biopanning were employed against Gaq using the standard method, and
RAPID flow cytometry was used to monitor the enrichment progression (Fig 15a). A late enrichment is observed where the maximum NMFI (2.36) is reached in round 5. Interestingly, an increase in SD was also observed which would suggest that the high-affinity binders also exhibit superior expression/display propensities (Fig 15b, Fig 9). In line with the findings from the CHIP biopanning campaign, where significant enrichment was achieved, RAPID FACS was not necessary. BIAS was employed to 95 clones from round 5, where 27 hits were identified (28% hit rate) (Fig 6, Table 4). The clones occupying the top 12 ranks were determined to be identical. Additionally, independent of these, clones ranked 13 to 16 were also found to be identical (Fig 15c). Identical clones exhibited similar BIAS koff values, and as demonstrated by the CHIP biopanning campaign, the candidate hit Fabs, which were biochemically characterized, displayed comparable koff values to those predicted by BIAS. Seven hits were predicted to be low expressors (flagged with*) and are ranked low on the BIAS rankings. This agrees with the RAPID flow cytometry results, where it was predicted that higher- affinity binders would also exhibit higher expression levels.
Table 4. BIAS hit summary table for Gotq. Inaccurate koff measurements are flagged (*- low expression #-unsaturuated Assoc- 1, !-poor exponential fit for Dissoc). Kd min and max are estimated based on general kou of previously discovered Craik lab Fabs (5 x 104 - 3 x 105)
Figure imgf000068_0001
RAPID biopanning identifies rare binders against CS3D
Cyclic STAT3 decoy (CS3D) is a dsDNA decoy that targets STAT3 and inhibits expression of downstream STAT3 induced genes17 18. To date, only a few recombinant Abs have been developed against nucleic acid targets, with most of them targeting unique structural motifs5,6. The identification of Fabs against CS3D, a 15-mer dsDNA target with no distinct structural motifs, is presented as an example of a challenging target for which limited Ab discovery precedence exists. Both standard biopanning and RAPID biopanning were performed in parallel to provide a head-to-head comparison of the two methods.
With the standard method, a total of six rounds of biopanning was performed with increasing stringency of washes and decreasing amounts of Ag per round. 95 colonies were randomly picked each round 4-6 (285 clones total) for candidate hit screening using dot blots. Four unique Fabs (3B11, 3B12, 3C8, and 4E4) were identified from 38 randomly sequenced clones, where three of the Fabs (3B11, 3B12, and 3C8) differed by only one amino acid indicating that a strong enrichment had occurred for a specific family of related clones over others throughout the rounds. Purified Fabs exhibited weak binding against CS3D with koff values ranging from 7.64 xlO'2 - 2.77 xlO'1 s'1 at 2 mM Fab concentrations (Table 5).
Table 5. koff (measured at 2pM) and recombinant expression yields (E. coli) of Fabs from CS3D biopanning.
Figure imgf000069_0001
To thoroughly examine candidate clones that were picked from the standard method campaign, BIAS was employed to the same clones picked previously that were randomly sequenced. BIAS resulted in a hit rate of 230 out of 285 clones (81% hit rate) (Table 6). However, when the top 10 ranked candidates from each round where sequenced, they were revealed to be the same low-affinity clones that were identified previously (Table 7), with two additional weak binders 2C11 (koff, 4.19 xlO'1 s'1) and 2B12 (koff, 3.7 xlO'1 s'1) being identified. . It was hypothesized that strong growth/display propensity biases were a dominant factor during the campaign, leading primarily to the identification of low affinity binders.
Table 6. BIAS of randomly chosen clones from Round 4-6 of panning and Round 4 Ph AB sorting.
Figure imgf000070_0001
Table 7. Top 10 BIAS ranked candidate clones from standard biopanning Round 4-6 and their koffS measured by BLI at 2 pM. Identical clones are italized or shown in bold.
Figure imgf000070_0002
RAPID biopanning was employed to identify higher affinity binders which was not achievable using the standard method. RAPID flow cytometry results showed that nearly no enrichment of binding had occurred during the campaign despite titers of output phage showing enrichment in round 3-5 (534-fold increase in output titer from round 5 relative to that of round 3), as shown with no significant changes of normalized MFI in any of the rounds (Fig 16a and Fig 7b). However, a higher fluorescent signal that does not significantly overlap with the control distribution (i.e., a minor shoulder distribution) emerged as biopanning proceeded, showing a maximum MFI at round 4. (Fig 16b and Fig. 7a). RAPID FACS was employed to isolate this population.
Beads from the round 4 output (top -4% fluorescent intensity) were sorted along with the beads from the majority population (bottom -96%) and both populations were used to infect TGI (E. coli) cells (Fig 16c). BIAS was performed separately against both populations where ~2-fold lower average BIAS koffS were observed in the top -4 percent population hits compared to the bottom -96 percent population hits (P< 0.001), suggesting that the top -4 percent population contained ph-Fab that have more favorable kinetic properties (Fig 16d). BIAS hits of the top -4 percent sorted population were sequenced, and a new candidate binder, SP1-B3, was identified, along with identical clones of previously identified low-affinity binders. SP1-B3 exhibited 2.11 xlO'2 s’1 koff which is ~3-fold lower than the previously identified Fab with the lowest kOff, and over -20- fold lower than the Fab with the highest kOff that was most frequently identified (Table 5). It is important to note that Fabs that were identified without RAPID biopanning exhibited significantly higher expression levels, as high as 27-fold (Table 5). As it has been well reported that the expression levels of displayed protein correlate with display propensities19 it is reasonable to conclude that display propensity bias was a strong factor during the biopanning campaign of CS3D, which led to the enrichment of weakly binding but high expressing ph-Fabs. The use of RAPID in this biopanning campaign enabled the direct isolation of a lower frequency, high affinity binder population, which resulted in the discovery of superior binding Abs, that otherwise would have not been discovered with the standard method.
Summary
The efficient identification of rare high-affinity Abs against challenging targets (i.e., low thermal stability, conformationally diverse, low expression of target, low antigenicity, low solubility, etc.) continues to be a major challenge in the Ab discovery field. For phage display based biopanning methods, challenging targets often exhibit weak Ab-Ag enrichment due to the low prevalence of high- affinity binders, and additional factors such as growth biases and inconsistent protein display propensities can affect, and even dominate the enrichment process. This ultimately leads to high-affinity binders never fully enriching and becoming difficult to identify, given the low probability of identification from common stochastic hit-picking screening methods.
The RAPID biopanning method was specifically designed as a solution for identifying rare high-affinity Abs against challenging targets using phage display. Where previous standard methods employ an approach where the total population of enriched displayed Abs are non- discriminatory screened, RAPID biopanning isolates/identifies a selective population of high affinity binders that are subsequently screened in a discriminatory manner. This was achieved by (1) accurately identifying the most enriched population of ph-Fab, (2) increasing the prevalence of low frequency high-affinity ph-Fabs by fluorescent activated sorting, and (3) rapidly screening candidate hits in a discriminatory matter to prioritize in-depth biochemical characterizations of promising binders. A method was developed for fluorescent labeling ph-Fab for quantitative measurements of ph-Fab bounding to Ag immobilized beads and a novel BLI method, BIAS, was developed for rapid real time analysis of candidate binders. Ultimately, RAPID biopanning follows a Label-Profile-Sort-Screen pipeline (Fig 1) and has been applied to two targets, CS3D and CHIP, where rare high-affinity binders were identified.
Robust labeling of ph-Fab was achieved using NHS-FITC. The labeling reaction is simple (only NHS-FITC and borate buffer required), fast (total reaction time 1 h in RT), reliable, and effective for the quantitative measurement of ph-Fab bound to Ag-Beads in flow cytometry. Most importantly, FITC labeling does not disrupt the binding of displayed Fab to Ag, nor does it exhibit any significant variabilities in labeling between different ph-Fabs.
By individually FITC labeling ph-Fab libraries from a single biopanning campaign and analyzing bound ph-Fab on Ag-Beads on flow cytometry, the enrichment progression of the campaign can be profiled. The progression of the global distribution of bound ph-Fab from subsequent rounds of biopanning can indicate dominant factors that affected enrichment (i.e., Fab-Ag binding, growth rates of phagemid containing E. coli. Fab display propensities). This is superior to phage ELIS As and calculations of phage titers of output ph-Fabs as these methods yield a single measurement of bound phage as opposed to a global distribution.
For campaigns that struggle to identify higher- affinity binders, fluorescent activated sorting of higher fluorescent populations can be an effective solution, as beads that exhibit higher fluorescent intensities contain greater enriched populations of higher- affinity binders. The frequency and variation in affinity of the higher- affinity binder can result in either a broader fluorescence distribution or a discernible population in FACS, as observed in the CS3D biopanning campaign. As shown with the CS3D biopanning campaign, a rare higher affinity binder, SP1-B3, was identified by isolating the ph-Fab-Ag-bead complexes exhibiting higher fluorescent signal by FACS. It is worth noting that previous efforts of standard magnetic bead biopanning followed by random clone picking or even applying the more stringent BIAS screening to these clones failed to identify new higher-affinity binders, indicating that the pool of ph-Fab examined itself had severely low frequencies of higher- affinity ph-Fabs. In the case for the CS3D campaign, the significantly lower expression levels of SP1-B3 (as much as ~20-fold less) compared to the weaker binders suggests that, at least in part, Fab display propensities played a role in the enrichment process. The example of the CS3D biopanning campaign highlights the use of fluorescent labeled ph-Fab libraries coupled with FACS, where rare high- affinity binders can be retrieved from a large pool of weaker binders, previously enriched for factors other than Fab-Ag binding.
It is crucial to the outcome of the antibody discovery campaign to correctly prioritize candidate binders for in-depth biochemical characterization as time and resources are practically limited in a lab setting. To this point a discriminatory hit screening method, BIAS, was developed to use Abs from crude extracts to measure real-time binding to immobilized Ags using BLI. The raw data is analyzed with an in-house developed script, BATCH, and clones are categorized and rank ordered based on their kinetic off-rates. The discriminatory identification of clones allows for the prioritization of more promising candidates and further investigation and characterizations can be done in line with the BIAS rankings. As shown with the CHIP biopanning campaign, BIAS is more discriminatory compared to conventional screening methods such as dot blots and provides more in-depth details of candidate clones compared to ELISAs (i.e., Fab expression levels, kOff values etc.). This is demonstrated by the most promising candidate binder, Fl, showing no obvious indication of being a promising candidate from dot blot results (Fig 5d). BIAS hits from the CHIP campaign show similar kinetic off-rate trends compared to the BIAS rankings, and of the five Fabs characterized, four show good agreement (~2-fold) of predicted koff values compared to biochemically characterized koffs.
RAPID biopanning, particularly where stringent fluorescent activated sorting was needed, can on occasion yield lower expression levels as shown with SP1-B3 (Table 5) or be poorer growers. Expression of these binders in full-length IgG formats in mammalian cells can help overcome low expression yields in bacteria, which is a widely employed solution in improving Ab expression. As an example, SP1-B3 expressed in an IgG format shows a 220-fold increase in expression (33 mg/L culture) compared to its Fab counterpail.
While the RAPID biopanning pipeline synergizes high quality ph-Fab population isolation with rapid discriminatory screening of candidate clones, each step is highly modular and can be utilized independently or in combination with other existing protocols. For example, as is common with YSD coupled with FACS, iterative rounds of fluorescent activated sorting could be performed for continuous enrichment of high quality ph-Fab populations. Also, where functional assays are available for hit screening, functional screens can be utilized in place of BIAS, post flow cytometry profiling, and/or fluorescent activated sorting. Finally, BIAS can be utilized with different Ab formats (i.e., scFv, nanobodies etc.) and different anti-tag secondary IgGs as well.
In conclusion, it is demonstrated that RAPID biopanning couples efficiency with precision to allow for a more selective biopanning campaign to identify rare high-affinity candidate clones, particularly for challenging targets.
Materials and Methods
Standard biopanning
Standard biopanning with magnetic beads were performed as previously described20,21. Briefly, a human naive B-cell phage displayed Fab library (diversity 4.1 xlO10) was used against CHIP and CS3D. Biotinylated Ags (EZ-link biotinylation Pierce) were immobilized to magnetic streptavidin beads (Dynabeads M-270 Streptavidin), and subsequently the ph-Fab library was added. For the CHIP campaign, 25 mM HEPES, 50 mM KC1 was used for the binding, and 25 mM HEPES, 50 mM KC1, 0.05% Tween-20 was used for washing. For the CS3D campaign, 20 mM Tris-HCl, 150 mM NaCl, 0.05% NP-40, 1 mM EDTA was used for the binding, and both PBS and PBS-T (0.05% Tween-20) was used for washing. Both campaigns introduced negative selections against magnetic beads with no Ag starting at round 2. Individual clones were selected and screened with either Dot blots, ELISAs, and BIAS and subsequent hits were sequenced. Plasmids containing unique sequences of Fabs were used to transform BL21(DE3) E. coli cells for further biochemical analysis of Fab. Phage preparation
Fab-display cd phage were first amplified and prepared using standard methods. Briefly, 50 ml cultures (2xYT, 2% glucose, 100 ug/ml Ampicillin) of phagemid containing E. coli cells were incubated at 37°C with 200 rpm shaking until OD600 reached ~0.5. Subsequently, 10 ml of this culture was infected with M13KO7 helper phage at 10:1 helper phage to cell ratio. Culture was incubated at 37°C for 30 min without shaking followed by 20 min with shaking at 200 rpm. Infected cells were collected by centrifugation and cells were resuspended with fresh media (2xYT 100 mg/ml Ampicillin, 50 mg/ml Kanamycin). Cultures were grown overnight, and amplified phage were isolated by adding PEG 6000/2.5M NaCl phage to the supernatant of the overnighted culture. Phage yield was analyzed by taking OD268 measurements.
Phage labeling with NHS-FITC
NHS-Fluorescein (Thermo Scientific) was prepared in DMSO (1 mg/ml final concentration). For the standard protocol labeling reaction consisted of the following: 50 pl of NHS-FITC (Img/ ml) stock 500 pl of Phage (OD268 = 1, final), and 40 pl of Borate buffer (0.67M). Previous studies have shown that NHS-esters, including NHS-FITC, can efficiently label M13 phage via the N-termini and lysine residues of the pVIII coat proteins22,2 . Labelling reaction was incubated in RT for 1 hr in the dark. PEG 6000/2.5M NaCl was added to the reaction to precipitate phage and incubated on ice for 15 min. Precipitated phage were collected by centrifugation (max speed for 5 min) and supernatant was removed. Pellets were resuspended in PBS and process was repeated. Phage were washed and precipitated three times total. Samples were immediately used in subsequent experiments. A plate reader (BioTek Synergy H4 plate reader) was used for fluorescent measurements and optical density of phage and FITC were analyzed using nanodrop. Phage labeling was aimed to maximize fluorescent signal per phage while maintaining solubility to achieve maximum dynamic range of fluorescent distribution. Labeling conditions were optimized using M13 helper phage. Normalized fluorescence shows an increase in relation to the number of FITC conjugated per phage, (Fig 12b). Further labeling caused precipitation and was not further investigated. The washing and the reaction incubation time was also optimized (Fig 12a, 12c) where a final reaction time of 1 hour and a total of three washes determined to be optimal. Flow cytometry and fluorescent activated sorting
All experiments of flow cytometry biopanning profiling and fluorescent activated sorting were done with biotinylated Ags immobilized with streptavidin coated beads. SPHEROTM Streptavidin Polystyrene particles, (3.0 - 3.9 pm) were used to immobilize Ag. First, the beads were blocked with 2% BSA- buffer for 1 hour. Beads were then washed by resuspending beads in 1 % BSA-buffer and subsequently centrifuging the beads (7k rpm for 2 min) to remove supernatant. This process was repeated three times. Biotinylated Ag was then added to beads at 1% BSA final concentration and incubated for one hour. Meanwhile, labeled phage were blocked in 1% BSA PBS for one hour. After Ag-immobilized beads were washed, blocked phage was added to the beads and incubated for 1-1.5 hours. After the binding phase, beads were washed 1-3 times with l%BSA-buffer and passed through a 40 pm cell strainer. Subsequently, flow cytometry analysis or fluorescent activated sorting was performed. All sorting was performed on a BDFACS Aria II and all flow cytometry analysis were performed on a benchtop Beckman Cytoflex Analyzer or BDFACSCaliber machine. Bead populations were gated with FSC and SSC parameters and later only singlet populations were analyzed by gating linear FSA and FSW. Of singlet population of beads, histogram analysis was done with FITC. For fluorescent activated sorting, correct percentage of events were sorted. Sorted beads were used to infect fresh TGI cells (OD600-0.7) and cells were plate for picking individual clones.
96-well Periylasmic extract preparation
Single colony clones were picked and inoculated into 2xYT media containing 2% glucose and 100 mg/ml Ampicillin in round bottom 96 well plates (150 ml of media per well). Cultures were grown overnight at 37°C with 200 rpm shaking. Following day 96 well cultures were inoculated (12 pl per well) into 96 well deep plates containing 2xYT media with 0.1% glucose and 100 pg/ml Ampicillin (1200 pl of media per well) and grown for 4-6 hours until culture is turbid. Fab expression was induced with 300 pl of 2xYT, ampicillin mg/ml and 5 mM IPTG and were left overnight at 30°C with 200 rpm shaking. Periplasmic extracts were collected by osmotic shock. Briefly, cells from overnight cultures were collected by centrifugation at 2000 g for 25 min and 375 pl of ice-cold TES buffer (200 mM Tris-HCl, 0.5 mM EDTA, 500 mM Sucrose, pH = 8) was added directly into each well and incubated with shaking at RT. Subsequently 1125 l of ice-cold water was added in each well and mixed thoroughly. The periplasmic fraction (supernatant) was collected by centrifugation at 2000 g for 25 min and stored at -20 °C for future experiments.
Dot blots
From 96-well periplasmic extracts, 2-3 pl was applied to nitrocellulose membranes. After ~10 min, the membrane was blocked with 2%-TBS-milk and gently rocked at RT. After 1 hr, membrane was gently rinsed with TBS-T (0.05% Tween-20) and 1%TBS-Milk with anti-myc HRP(9E10) (1:5000 dilution) was added and incubated for 1 hr. Washes were performed with TBS-T (x2) and TBS (x2), and 1 ml of Immobilon Forte Western HRP substrate was added for imaging on a ChmiDoc MP imaging system.
Biolayer interferometry (BLI)
All buffers were filter sterilized with 0.22 mm filters prior to preparing samples. Black 384 well microplates were used to set up BLI plates and streptavidin tips were purchased by Sartorius. Kinetic constants of Fabs were determined by Octet RED384 system with continuous shaking at 1000 rpm in RT. Prior to the experiment, biological tips (model name) were presoaked in buffer to allow for equilibration for 1 hour. Data were analyzed using 1 : 1 interaction model on the ForteBio data analysis software 12.0.
Biolayer interferometry antibody screen (BIAS)
Instrument and reagent set up was identical to that of standard BLI. Anti-myc Ab (9E10) (Merk) was used for the Assoc-2. For the control experiments, BLI was run in following method: Baseline (1 min), Load, Baseline (1 min), Assoc-1 (10 min), Assoc-2 (10 min), Dissoc (10 min), and for CHIP and CS3D BIAS runs, Assoc-1 (3 min), Assoc-2 (2 min 30 sec), and Dissoc (3 min) was used as the final protocol. Dependent on the Ag that was used, loading step was adjusted where loading was immediately terminated once rate of loading changed to allow for equal distribution of Ag on tip. Raw data files of the run(s) were then used in the BATCH to rank order candidate hits according to their predicted dissociation rates. The true Assoc-2 slope of the PPE spiked sample without 9E10 calculated by BATCH was further used as the threshold value for determining hits (Table 1). BIAS Algorithm Triaging Confirmed Hits (BATCH) development.
A BIAS function was developed to effectively process raw BLI sensogram data, to categorize and rank order candidate hits according to their kinetic properties. A brief schematic of the categorization, ranking system, and outputs is listed in Fig. 10. BATCH will process all raw BLI data files, a user-created methods file, and a user-created thresholds file. Due to buffer mismatches occurring during the BIAS runs, each step is trimmed at the beginning and end to avoid erroneous noise. Briefly, the Assoc- 1 and Dissoc steps are fitted as a one phase exponential association and one phase exponential decay respectively. To account for continued Fab binding during the Assoc-2 step, the one phase exponential association fit of the Assoc- 1 step is extrapolated through Assoc-2 as the “extrapolated association- 1 curve”. The “true Assoc- 2 slope” only accounts for the anti-myc IgG signal contribution and is calculated by subtracting the extrapolated association curve by the raw Assoc-2 curve. The “true Assoc-2 slope” is fitted with a linear regression to quantify a significant signal shift. Importantly the “true Assoc-2 slope” distinguishes “Hits” versus “False positives”. Threshold values were determined by control experiments (Fig 4b-4d, Table 1). BATCH outputs R2 values for all fits in each step which determines which category each clone is classified into based on a threshold value (figure 11). BATCH assumes a pseudo first-order kinetic model for predicting koffs. By using the calculated koffs from the Dissoc step, BATCH will predict a range of Kd based on previous Craik lab Fab kons and rank clones classified as “Hits”. BATCH will generate two tables to provide a summary of results and a more detailed view of all processed data. To give the user more information on the reason behind classification of clones, the comment section provides more details (i.e., no dissociation, no association, R2 values too low etc.). Threshold and kon values can be changed according to the user’s experimental set up where secondary IgG or Ab format is different.
Exceptions to the BIAS rankings
Clones that are flagged in the BIAS rankings indicate inaccuracies in koff predictions. For these clones, the “true association-2 slope” can be a better predictor of binding, as exchange rates of Fab to Fab-9E10 complex during Assoc-2 are directly correlated to koff of the Fab. Specifically, rapid exchange of Fab to Fab-9E10 indicate a high koff, which translate to a high “true association-2 slope”. Therefore, koff is inversely proportional to the true association-2 slope. A simple guide to further distinguishing flagged BIAS results is shown in Fig 13. Fab expression
Freshly transformed BL21(DE3) E. coli single colony clones were picked and inoculated into 50 ml of 2xYT media containing 2% glucose and 100 mg/ml Ampicillin. Starter cultures were grown overnight at 37oC with 200 rpm shaking. The next day, the starter culture was inoculated to 1 L of 2xYT media with 0.1% glucose and 100 mg/ml Ampicillin (OD600 0.05 final) and after incubation (37°C with 200 rpm shaking), protein expression was induced with IPTG (1 mM final) at OD600 of 0.6 and continued to grow overnight (20°C with 200 rpm shaking). Periplasmic extracts containing Fabs were collected by osmotic shock. Briefly, cells from overnight cultures were collected by centrifugation at 9000 g for 15 min and -15 ml of ice-cold TES buffer was added and resuspended thoroughly to achieve a homogenous mixture. The mixture was then incubated with gentle shaking at 4oC for 1 hour. Subsequently -25 ml of ice-cold water was added and incubated with gentle shaking for 45 min. The periplasmic fraction (supernatant) was collected by centrifugation at 10,000 g for 30 min and loaded to Ni- NTA resin for affinity purification. Purified Fabs were dialyzed against PBS (lOkDa MWCO) and size exclusion chromatography was performed to further purify the Fabs and remove any aggregates (AKTA autopurification system with a Superdex 200 10/300GL column). Fractions exhibiting correct size were pooled and Fab concentrations were determined by absorbance at 280 nm.
Conversion of Fab into Full-Length IgG
The methods used here were described in detail in a previous study 24. Briefly, the heavy and light chain regions of Fabs were cloned out from their respective phagemids and then individually cloned into a pTT5-SP-Hl mammalian expression vector through Gibson Assembly® (NEB #E2611) methods. The Gibson assembly product was transformed into NEB® 5a Competent E. Coli cells (NEB #C2987H) and plasmids were isolated using a ZymoPurell Plasmid Maxiprep Kit (Zymo Research #D4203) and confirmed through sequencing.
Small-scale Expression and Purification of Full-Length IgG in Mammalian Cells
The expression protocol used here was based on the Expi293TM Expression System (ThermoFisher #A14635). Expi293 suspension cells were seeded at a final density of 2.5 x 106 cells/mL in a 6-well plate. The next day cells were diluted to a final density of 3 x 106 cells/mL and a total of 1 .0 pg/mL of plasmid DNA was transfected using ExpiFectamineTM 293 Transfection Kit. Transfected cells were then incubated on an orbital shaker at 37°C and 8% CO2 overnight and ExpiFectamine 293 Transfection Enhancer 1 and 2 were added to the cells. Secreted IgG was harvested 6 days post-transfection and the supernatant was incubated with PierceTM Protein G Plus Agarose (ThermoScientific #22851) resin at room temperature for 1 hour. After washing with 10 column volumes of PBS, the IgG was eluted with 2 column volumes of 100 mM glycine, pH 2.5 and neutralized by 1 M Tris, pH 8.5. The eluted fractions were determined by SDS-PAGE gel and fractions containing pure IgG were collected and dialyzed against PBS buffer.
Statistics and Reproducibility
NHS-FITC phage labeling experiments were performed in triplicates and data were presented as mean ± SD. Flow cytometry and FACS data was analyzed by FlowJo 10.9.0 from at least 10,000 events. The statistical significance between the BIAS off-rates of sorted and unsorted populations were determined by A two-way ANOVA test on Prism (Graphpad).
References
1. Basu, K., Green, E. M., Cheng, Y. & Craik, C. S. Why recombinant antibodies — benefits and applications. Pharm. Biotcchnol. • Chcm. Biotcchnol. 60, 153-158 (2019).
2. Asarnow, D. et al. Structural insight into SARS-CoV-2 neutralizing antibodies and modulation of syncytia. Cell 184, 3192-3204.el6 (2021).
3. Taylor, P. C. et al. Neutralizing monoclonal antibodies for treatment of CO VID- 19. Nat. Rev. Immunol. 21, 382-393 (2021).
4. Miersch, S. et al. Ultrapotent and broad neutralization of SARS-CoV-2 variants by modular, tetravalent, bi-paratopic antibodies. Cell Rep. 39, 110905 (2022).
5. Zeraati, M. et al. I-motif DNA structures are formed in the nuclei of human cells. Nat. Chem. 10, 631-637 (2018).
6. Ye, J.-D. et al. Synthetic antibodies for specific recognition and crystallization of structured RNA. Proc. Natl. Acad. Sci. U. S. A. 105, 82-87 (2008).
7. Fellouse, F. A. et al. High-throughput Generation of Synthetic Antibodies from Highly Functional Minimalist Phage-displayed Libraries. J. Mol. Biol. 373, 924-940 (2007).
8. Jones, M. L. et al. Targeting membrane proteins for antibody discovery using phage display. Sci. Rep. 6, 26240 (2016).
9. Koide, A., Wojcik, J., Gilbreth, R. N., Hoey, R. J. & Koide, S. Teaching an old scaffold new tricks: monobodies constructed using alternative surfaces of the FN3 scaffold. J. Mol. Biol. 415, 393-405 (2012). 10. Wellner, A. et al. Rapid generation of potent antibodies by autonomous hypermutation in yeast. Nat. Chem. Biol. 17, 1057-1064 (2021).
11. Lopez Tyler et al. Functional selection of protease inhibitory antibodies. Proc. Natl. Acad. Sci. 116, 16314-16319 (2019).
12. Boder, E. T. & Wittrup, K. D. Yeast surface display for screening combinatorial polypeptide libraries. Nat. Biotechnol. 15, 553-557 (1997).
13. VanAntwerp, J. J. & Wittrup, K. D. Fine affinity discrimination by yeast surface display and flow cytometry. Biotechnol. Prog. 16, 31-37 (2000).
14. Hilpert, K. et al. Anti-c-myc antibody 9E10: epitope key positions and variability characterized using peptide spot synthesis on cellulose. Protein Eng. Des. Sei. 14, 803-806 (2001).
15. Ravalin, M. et al. Specificity for latent C termini links the E3 ubiquitin ligase CHIP to caspases. Nat. Chem. Biol. 15, 786-794 (2019).
16. Ng, S. et al. Discovery and Structure-Based Design of Macrocyclic Peptides Targeting STUB1. (2021) doi:10.26434/chemrxiv-2021- 1108b.
17. Sansone, P. & Bromberg, J. Targeting the interleukin-6/Jak/stat pathway in human malignancies. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 30, 1005-1014 (2012).
18. Johnson, D. E., O’Keefe, R. A. & Grandis, J. R. Targeting the 1L-6/JAK/STAT3 signalling axis in cancer. Nat. Rev. Clin. Oncol. 15, 234-248 (2018).
19. Scott, N. et al. Single-chain Fv phage display propensity exhibits strong positive correlation with overall expression levels. BMC Biotechnol. 8, 97 (2008).
20. Duriseti, S. et al. Antagonistic anti-urokinase plasminogen activator receptor (uPAR) antibodies significantly inhibit uPAR-mediated cellular signaling and migration. J. Biol. Chem. 285, 26878-26888 (2010).
21. Kim, J. M., Stroud, R. M. & Craik, C. S. Rapid identification of recombinant Fabs that bind to membrane proteins. Methods 55, 303-309 (2011).
22. Yacoby, I., Shamis, M., Bar, H., Shabat, D. & Benhar, I. Targeting antibacterial agents by using drug-carrying filamentous bacteriophages. Antimicrob. Agents Chemother. 50, 2087-2097 (2006).
23. Li, K. et al. Chemical Modification of M13 Bacteriophage and Its Application in Cancer Cell Imaging. Bioconjug. Chem. 21, 1369-1377 (2010).
24. Duriseti, S. et al. Antagonistic anti-urokinase plasminogen activator receptor (uPAR) antibodies significantly inhibit uPAR-mediated cellular signaling and migration. J. Biol. Chem. 285, 26878-26888 (2010).
25. Van Raamsdonk, C. D. et al. Frequent somatic mutations of GNAQ in uveal melanoma and blue naevi. Nature 457, 599-602 (2009).
26. Shirley, M. D. et al. Sturge-Weber Syndrome and Port-Wine Stains Caused by Somatic Mutation in GNAQ. N. Engl. J. Med. 368, 1971-1979 (2013). In at least some of the previously described embodiments, one or more elements used in an embodiment can interchangeably be used in another embodiment unless such a replacement is not technically feasible. It will be appreciated by those skilled in the art that various other omissions, additions and modifications may be made to the methods and structures described above without departing from the scope of the claimed subject matter. All such modifications and changes are intended to fall within the scope of the subject matter, as defined by the appended claims.
It will be understood by those within the ail that, in general, terms used herein, and especially in the appended claims (e.g., bodies of the appended claims) are generally intended as “open” terms (e.g., the term “including” should be interpreted as “including but not limited to,” the term “having” should be interpreted as “having at least,” the term “includes” should be interpreted as “includes but is not limited to,” etc.). It will be further understood by those within the art that if a specific number of an introduced claim recitation is intended, such an intent will be explicitly recited in the claim, and in the absence of such recitation no such intent is present. For example, as an aid to understanding, the following appended claims may contain usage of the introductory phrases “at least one” and “one or more” to introduce claim recitations. However, the use of such phrases should not be construed to imply that the introduction of a claim recitation by the indefinite articles “a” or “an” limits any particular claim containing such introduced claim recitation to embodiments containing only one such recitation, even when the same claim includes the introductory phrases “one or more” or “at least one” and indefinite articles such as “a” or “an” (e.g., “a” and/or “an” should be interpreted to mean “at least one” or “one or more”); the same holds true for the use of definite articles used to introduce claim recitations. In addition, even if a specific number of an introduced claim recitation is explicitly recited, those skilled in the art will recognize that such recitation should be interpreted to mean at least the recited number (e.g., the bare recitation of “two recitations,” without other modifiers, means at least two recitations, or two or more recitations). Furthermore, in those instances where a convention analogous to “at least one of A, B, and C, etc.” is used, in general such a construction is intended in the sense one having skill in the art would understand the convention (e.g., “ a system having at least one of A, B, and C” would include but not be limited to systems that have A alone, B alone, C alone, A and B together, A and C together, B and C together, and/or A, B, and C together, etc.). In those instances where a convention analogous to “at least one of A, B, or C, etc.” is used, in general such a construction is intended in the sense one having skill in the art would understand the convention (c.g., “ a system having at least one of A, B, or C” would include but not be limited to systems that have A alone, B alone, C alone, A and B together, A and C together, B and C together, and/or A, B, and C together, etc.). It will be further understood by those within the art that virtually any disjunctive word and/or phrase presenting two or more alternative terms, whether in the description, claims, or drawings, should be understood to contemplate the possibilities of including one of the terms, either of the terms, or both terms. For example, the phrase “A or B” will be understood to include the possibilities of “A” or“B” or “A and B.”
In addition, where features or aspects of the disclosure are described in terms of Markush groups, those skilled in the art will recognize that the disclosure is also thereby described in terms of any individual member or subgroup of members of the Markush group.
As will be understood by one skilled in the art, for any and all purposes, such as in terms of providing a written description, all ranges disclosed herein also encompass any and all possible sub-ranges and combinations of sub-ranges thereof. Any listed range can be easily recognized as sufficiently describing and enabling the same range being broken down into at least equal halves, thirds, quarters, fifths, tenths, etc. As a non-limiting example, each range discussed herein can be readily broken down into a lower third, middle third and upper third, etc. As will also be understood by one skilled in the art all language such as “up to,” “at least,” “greater than,” “less than,” and the like include the number recited and refer to ranges which can be subsequently broken down into sub-ranges as discussed above. Finally, as will be understood by one skilled in the art, a range includes each individual member. Thus, for example, a group having 1-3 articles refers to groups having 1, 2, or 3 articles. Similarly, a group having 1-5 articles refers to groups having 1, 2, 3, 4, or 5 articles, and so forth.
Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, it is readily apparent to those of ordinary skill in the art in light of the teachings of this invention that certain changes and modifications may be made thereto without departing from the spirit or scope of the appended claims. Accordingly, the preceding merely illustrates the principles of the invention. It will be appreciated that those skilled in the art will be able to devise various arrangements which, although not explicitly described or shown herein, embody the principles of the invention and are included within its spirit and scope. Furthermore, all examples and conditional language recited herein arc principally intended to aid the reader in understanding the principles of the invention and the concepts contributed by the inventors to furthering the ail, and are to be construed as being without limitation to such specifically recited examples and conditions. Moreover, all statements herein reciting principles, aspects, and embodiments of the invention as well as specific examples thereof, are intended to encompass both structural and functional equivalents thereof. Additionally, it is intended that such equivalents include both currently known equivalents and equivalents developed in the future, i.e., any elements developed that perform the same function, regardless of structure. Moreover, nothing disclosed herein is intended to be dedicated to the public regardless of whether such disclosure is explicitly recited in the claims. The scope of the present invention, therefore, is not intended to be limited to the exemplary embodiments shown and described herein. Rather, the scope and spirit of present invention is embodied by the appended claims. In the claims, 35 U.S.C. § 112(f) or 35 U.S.C. §112(6) is expressly defined as being invoked for a limitation in the claim only when the exact phrase "means for" or the exact phrase "step for" is recited at the beginning of such limitation in the claim; if such exact phrase is not used in a limitation in the claim, then 35 U.S.C. § 112 (f) or 35 U.S.C. § 112(6) is not invoked.

Claims

CLAIMS WHAT IS CLAIMED IS:
1. A method for identifying an antibody with affinity to a target of interest by using biolayer interferometry (BLI), the method comprising: contacting a sensor tip comprising the target of interest immobilized thereon with a first aliquot of a solution comprising multiple copies of a first antibody; irradiating the sensor tip with an incident light and measuring a first reflected light, wherein a wavelength shift of first reflected light compared to the incident light is indicative of binding of the first antibody to the target of interest immobilized on the sensor tip; contacting the sensor tip with a second aliquot of the solution comprising multiple copies of the first antibody, wherein the second aliquot further comprises a second antibody that binds to the first antibody; and irradiating the sensor tip with the incident light and measuring a second reflected light, wherein a wavelength shift of second reflected light compared to the incident light and/or the first reflected light is indicative of binding of the second antibody to the first antibody bound to the sensor tip, wherein presence of: (a) the wavelength shift of first reflected light compared to the incident light and (b) the wavelength shift of the second reflected light compared to the incident light and/or the first reflected light identifies the first antibody as having affinity to the target of interest.
2. The method according to Claim 1, wherein the method further comprises contacting the sensor tip with a buffer and measuring a dissociation rate of the first antibody from the target of interest.
3. The method according to Claims 1 or 2, wherein the method is performed simultaneously or sequentially on a plurality of solutions comprising different antibodies.
4. The method according to any one of Claims 1 to 3, wherein the antibodies are produced from a phage display library.
5. The method according to any one of Claims 1 to 4, wherein the antibodies are Fab, Fv, scFv, VH domain antibodies, or nanobodies.
6. The method according to any one of Claims 1-5, wherein the solution comprising multiple copies of the first antibody is generated from a bacteria infected with a phage displaying the first antibody, wherein the phage is identified by a method comprising: fluorescently labeling a phage display library comprising a multitude of different antibodies displayed on phages; contacting the labeled phage display library with the target of interest immobilized on beads to produce bead-phage complexes; and analyzing the bead-phage complexes using flow cytometry to generate a profile of the complexes; using the profile to identify high-affinity antibodies.
7. The method according to Claim 6, wherein the profile comprises a histogram representing the frequency of complexes having different fluorescence intensities.
8. The method according to Claim 7, wherein a median fluorescence intensity (MFI) and/or a standard deviation (SD) are calculated for one or more components of the profile.
9. The method according to Claim 8, wherein the high affinity antibodies correspond to a component of the profile having a high MFI.
10. The method according to Claims 8 or 9, wherein the high affinity antibodies correspond to a component of the profile having a high SD.
11 . The method of according to any one of Claims 6 to 10, further comprising: sorting the bcad-phagc complexes based on a level of fluorescent phages bound to the beads, thereby separating complexes exhibiting a threshold level of fluorescence from complexes exhibiting fluorescence below the threshold level.
12. The method according to any one of Claims 6 to 11, wherein the phage display library comprises phages from two or more rounds of preselection for phages that bind to the target of interest.
13. The method according to Claim 12, wherein the phage display library comprises phages from four or more rounds of preselection for phages that bind to the target of interest.
14. The method according to Claims 12 or 13, wherein phages from each round are separately contacted with the target of interest immobilized on beads and analyzed using flow cytometry to generate a profile for phages from each round of preselection.
15. The method according to Claim 14, comprising using the generated profiles to identify high-affinity antibodies.
16. The method according to Claim 15, wherein the high-affinity antibodies have the profile with the highest MFI.
17. The method according to any one of Claims 6 to 16, wherein the phage display library is labeled with fluorescein isothiocyanate (FITC) or an Alexa Fluor (AF) dye.
18. The method according to Claim 17, wherein the phage display library is labeled with FITC.
19. The method according to Claim 17, wherein the phage display library is labeled with AF647.
20. The method according to any one of Claims 6 to 19, wherein the phages are M 13 phages.
21. The method according to Claim 20, wherein the M13 pages are labeled via N-termini and/or lysine residues of the pVIII coat proteins.
22. The method according to any one of Claims 6 to 21, wherein the phage display library is generated from bacteria infected with a phage.
23. The method according to Claim 22, wherein the bacteria are Escherichia coli.
24. The method according to Claims 22 or 23, wherein the solution comprising multiple copies of a first antibody is crude periplasmic extract (PPE) from bacteria infected with a phage.
25. The method according to Claim 24, wherein the first antibody comprises a tag and the second antibody binds to the tag.
26. The method according to any one of Claims 2-25, wherein the buffer comprises no detectable copies of the first antibody or the second antibody.
27. The method according to any one of Claims 2-26, wherein the method further comprises determining a binding kinetic parameter of first antibody to the target of interest using tip measurements.
28. The method according to Claim 27, wherein the binding kinetic parameter is a dissociation rate constant.
29. The method according to Claim 28, wherein the dissociation rate constant is determined using measurements generated while the tip is contacting the buffer.
30. The method according to any one of the preceding Claims, wherein the method further comprises determining false positives using tip measurements.
31. The method according to Claim 30, wherein the false positives arc determined using measurements generated while the tip is contacting the second aliquot of the solution comprising multiple copies of the first antibody and wherein the second aliquot further comprises the second antibody that binds to the first antibody, wherein presence of (a) and absence of (b) identifies the first antibody as a false positive antibody.
32. The method according to any one of the preceding Claims, wherein the method further comprises determining if the sensor tip is saturated by antibody copies using tip measurements.
33. The method according to Claim 32, wherein the saturation is determined using measurements generated while the tip is in the first aliquot of the solution.
34. A system configured to perform the method according to any one of Claims 1 to 33, optionally wherein the system comprises a biolayer interferometry device and a cell sorter.
35. A biolayer interferometry device configured to perform the screening according to any one of Claims 1 to 33.
36. A method for identifying antibodies with affinity to a target of interest, the method comprising: conducting a first round of biopanning comprising isolating phages displaying antibodies that bind to the target of interest and fluorescently labeling the phages to generate a first labeled population of phages; conducting a second round of biopanning comprising isolating phages displaying antibodies that bind to the target of interest and fluorescently labeling the phages to generate a second labeled population of phages; separately contacting the first and the second labeled population of phages with the target of interest immobilized on beads to produce bead-phage complexes; analyzing the complexes using flow cytometry to generate a first fluorescence profile of the complexes produced from the first labeled population of phages and a second fluorescence profile of second labeled population of phages; and using the first and second profiles to identify antibodies with affinity to the target of interest.
37. The method according to Claim 36, wherein the profile comprises a histogram representing the frequency of complexes having different fluorescence intensities.
38. The method according to Claim 37, wherein a median fluorescence intensity (MFI) and/or a standard deviation (SD) are calculated for one or more components of the profiles.
39. The method according to Claim 38, wherein the antibodies associated with the profile having the highest MFI are identified as high affinity antibodies.
40. The method according to Claim 38, wherein when the MFIs of each profile are similar, the antibodies associated with the profile with the highest SD are identified as high affinity antibodies.
41. The method according to any one of Claims 38 to 40, wherein the antibodies associated with the profile with the highest MFI and SD are identified as high affinity antibodies.
42. The method of according to any one of Claims 36 to 41, wherein the isolating further comprises: sorting the complexes based on a level of fluorescent phages bound to the beads, thereby separating complexes exhibiting a threshold level of fluorescence from complexes exhibiting fluorescence below the threshold level; wherein the high affinity antibodies are present in the complexes exhibiting the threshold level of fluorescence.
43. The method according to any one of Claims 36 to 42, wherein the method comprises conducting three or more rounds of biopanning to generate three or more labeled populations of phages.
44. The method according to Claim 43, wherein the method comprises conducting four or more rounds of biopanning to generate four or more labeled populations of phages.
45. The method according to any one of Claims 36 to 44, wherein the method further comprises screening the high affinity antibodies individually to identify antibodies with a high affinity to the target of interest using bio-layer interferometry (BLI).
46. The method according to Claim 45, wherein the screening comprises: infecting bacteria with phages expressing the high affinity antibodies; separating the bacteria into single cells and growing bacterial colonies that each express a single type of antibody; preparing an extract from the colonies to provide a plurality of solutions, each solution comprising multiple copies of single type of antibody; contacting in parallel, a first aliquot of each of the plurality of solutions with a sensor tip comprising the target of interest immobilized thereon; contacting in parallel, a second aliquot of each of the plurality of solutions with the sensor tip, wherein the second aliquot further comprises a second antibody that binds to each of the single type of antibodies; wherein binding between the target of interest, the antibodies expressed by the phages, and the second antibody are determined by irradiating the target of interest with light and measuring wavelength shift over time using the sensor tip; and wherein a difference in the rate of change of wavelength shift over time between when the sensor tip is in contact with first aliquot and when the sensor tip is in contact with the second aliquot is used to identify whether the antibody expressed by the phages has affinity to the target of interest.
47. The method according to Claim 46, wherein the method further comprises contacting a third buffer with the sensor tip.
48. The method according to Claims 46 or 47, wherein the phages produce an antibody comprising a tag and the second antibody binds to the tag.
49. The method according to any one of Claims 46 to 48, wherein the screening is performed for phages produced from at least 10, at least 30, at least 100, at least 300, or more individual bacterial colonies.
50. The method according to any one of Claims 36 to 49, wherein the phage display library is labeled with fluorescein isothiocyanate (FITC) or an Alexa Fluor (AF) dye.
51. The method according to Claim 50, wherein the phage display library is labeled with FITC.
52. The method according to Claim 50, wherein the phage display library is labeled with AF647.
53. The method according to any one of Claims 36 to 52, wherein the phages are M13 phages.
54. The method according to Claim 53, wherein the M13 pages are labeled via N-termini and/or lysine residues of the pVIII coat proteins.
55. The method according to any one of Claims 46 to 54, wherein the phage display library is generated using bacteria of the genus Escherichia.
56. The method according to Claim 55, wherein the bacteria are Escherichia coli bacteria.
57. The method according to any one of Claims 44 to 56, wherein the solution comprises crude pcriplasmic extract (PPE) produced from bacteria.
58. The method according to any one of Claims 47 to 56, wherein the buffer does not include a chaotropic agent.
59. The method according to any one of Claims 47 to 58, wherein the buffer comprises a chaotropic agent.
60. The method according to any one of Claims 47 to 59, wherein the method further comprises determining a binding kinetic parameter of the antibodies to the target of interest using tip measurements.
61. The method according to Claim 60, wherein the binding kinetic parameter is a dissociation rate constant.
62. The method according to Claim 61, wherein the dissociation rate constant is determined using measurements generated while the tip is in the buffer.
63. The method according to any one of Claims 46 to 62, wherein the method further comprises determining false positives using tip measurements.
64. The method according to Claim 63, wherein the false positives are determined using measurements generated while the tip is in the second aliquot.
65. The method according to any one of Claims 46 to 64, wherein the method further comprises determining if the sensor tip is saturated by antibody copies using tip measurements.
66. The method according to Claim 65, wherein the saturation is determined using measurements generated while the tip is in the first aliquot the solution.
67. A system configured to perform the method according to any one of Claims 36 to 66.
68. A biolayer interferometry device configured to perform the screening according to any one of Claims 35 to 66.
69. A system for identifying antibodies with affinity to a target of interest, the system comprising: a processor comprising memory operably coupled to the processor wherein the memory comprises instructions stored thereon, which when executed by the processor, cause the processor to: obtain raw bio-layer interferometry data, wherein the interferometry data comprises sequential wavelength shift measurements resulting from interactions between a sensor tip comprising a target of interest, copies of an antibody, and tag binding antibodies; section the interferometry data into two consecutive segments; fit a one phase exponential association curve to raw data from the first segment, wherein the sequentially later end of the one phase exponential association curve comprises a first slope; fit a line to the second segment data, wherein the fitted line comprises a second slope; and compare the first slope to the second slope, wherein the antibody copies are identified as having affinity to the target of interest if the second slope is greater than the first slope.
70. The system according to Claim 69, wherein the memory comprises instructions stored thereon, which when executed by the processor, cause the processor to refine raw data from the second segment by extrapolating the association curve fit to the first segment through the second segment and subtracting the extrapolated association curve from the raw data of the second segment, wherein the line is fit to the refined second segment data.
71 . The system according to Claims 69 or 70, wherein the memory comprises instructions stored thereon, which when executed by the processor, cause the processor to section the interferometry data into three consecutive segments, wherein a one phase exponential decay curve is fit to raw data from the third segment.
72. The system according to Claim 71, wherein the memory comprises instructions stored thereon, which when executed by the processor, cause the processor to determine a goodness of fit metric for each fitted curve.
73. The system according to Claim 72, wherein the goodness of fit metric is an r-squared value.
74. The system according to Claim 73, wherein the memory comprises instructions stored thereon, which when executed by the processor, cause the processor to detect negatives of the antibody copies not binding to the target of interest, false positives of non-specific binding proteins binding to the target of interest, and/or hits of the antibody copies binding to the target of interest.
75. The system according to Claim 74, wherein negatives and false positives are detected by determining if the slope of the line fit to the second segment is below a predetermined threshold value.
76. The system according to Claim 74 or 75, wherein negatives and false positives are detected by determining if the r-squared value of the line fit to the second segment is below a predetermined threshold value.
77. The system according to any one of Claim 74 to 76, wherein false positives are detected by determining if the r-squared value of the association curve fit to the first segment meets or exceeds a predetermined threshold value.
78. The system according to any one of Claim 74 to 76, wherein negatives are detected by determining if the r-squarcd value of the association curve fit to the first segment is below a predetermined threshold value.
79. The system according to any one of Claim 74 to 78, wherein the hits are detected by determining if the slope of the line fit to the second segment meets of exceeds a predetermined threshold value and if the r-squared value of the line fit to the second segment meets or exceeds a predetermined threshold value.
80. The system according to Claim 79, wherein the memory comprises instructions stored thereon, which when executed by the processor, cause the processor to determine if the sensor tip is saturated by antibody copies by calculating a slope of the extrapolated association curve.
81. The system according to Claim 80, wherein the sensor tip is determined to be saturated if the slope of the extrapolated association curve meets or exceeds a predetermined threshold value.
82. The system according to any one of the Claims 79 to 81, wherein the memory comprises instructions stored thereon, which when executed by the processor, cause the processor to determine antibody clone expression by calculating the total wavelength shift of the first segment.
83. The system according to Claim 82, wherein the antibody copies arc determined to have low expression if the total wavelength shift of the first segment is below a predetermined threshold value.
84. The system according to any one of the Claims 79 to 83, wherein the memory comprises instructions stored thereon, which when executed by the processor, cause the processor to predict a dissociation rate for the antibody copies and the target of interest.
85. The system according to Claim 84, wherein the dissociation rate is predicted using the exponential decay curve fit to the third segment.
86. The system according to Claims 84 or 85, wherein the memory comprises instructions stored thereon, which when executed by the processor, cause the processor to determine if the predicted dissociation rate is inaccurate.
87. The system according to Claim 86, wherein the dissociation rate is determined to be inaccurate if the r-squared value of the exponential decay curve fit to the third segment is below a predetermined threshold value.
88. The system according to any one of the Claims 69 to 87, wherein the system is configured to identify antibodies with affinity to the target of interest for a plurality of inputs each comprising sequential wavelength shift measurements resulting from interactions between a sensor tip comprising a target of interest, copies of an antibody, and tag binding antibodies.
89. The system according to Claim 88, wherein the copies for each input are of a different antibody.
90. The system according to Claims 88 or 89, wherein the memory comprises instructions stored thereon, which when executed by the processor, cause the processor to rank the inputs based on a binding kinetic parameter predicted for each of the different antibodies to the target of interest.
91. The system according to Claim 90, wherein the binding kinetic parameter is a dissociation rate constant.
92. The system according to any one of the Claims 69 to 91, wherein the system further comprises a bio-layer interferometry sensor tip configured to generate the raw bio-layer interferometry data and transmit it to the processor, the sensor tip comprising: a surface comprising the target of interest; a light source configured to irradiate the surface; and a detector configured to detect an interaction between antibodies and the target of interest.
PCT/US2024/0311362023-06-012024-05-24Rare antibody phage isolation and discrimination (rapid) biological panningWO2024249362A1 (en)

Applications Claiming Priority (4)

Application NumberPriority DateFiling DateTitle
US202363505665P2023-06-012023-06-01
US63/505,6652023-06-01
US202363584750P2023-09-222023-09-22
US63/584,7502023-09-22

Publications (1)

Publication NumberPublication Date
WO2024249362A1true WO2024249362A1 (en)2024-12-05

Family

ID=93658386

Family Applications (1)

Application NumberTitlePriority DateFiling Date
PCT/US2024/031136WO2024249362A1 (en)2023-06-012024-05-24Rare antibody phage isolation and discrimination (rapid) biological panning

Country Status (1)

CountryLink
WO (1)WO2024249362A1 (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication numberPriority datePublication dateAssigneeTitle
US6289144B1 (en)*1995-05-122001-09-11Novartis AgSensor platform and method for the parallel detection of a plurality of analytes using evanescently excited luminescence
US20050254062A1 (en)*2003-11-062005-11-17Fortebio, Inc.Fiber-optic assay apparatus based on phase-shift interferometry
US20200306374A1 (en)*2017-07-112020-10-01Compass Therapeutics LlcAgonist antibodies that bind human cd137 and uses thereof
WO2021207433A2 (en)*2020-04-072021-10-14The Regents Of The University Of CaliforniaEpitopes of sars-cov-2 neutralizing antibodies
US20220289866A1 (en)*2019-07-122022-09-15The Regents Of The University Of CaliforniaChemically controlled monoclonal antibody target engagement

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication numberPriority datePublication dateAssigneeTitle
US6289144B1 (en)*1995-05-122001-09-11Novartis AgSensor platform and method for the parallel detection of a plurality of analytes using evanescently excited luminescence
US20050254062A1 (en)*2003-11-062005-11-17Fortebio, Inc.Fiber-optic assay apparatus based on phase-shift interferometry
US20200306374A1 (en)*2017-07-112020-10-01Compass Therapeutics LlcAgonist antibodies that bind human cd137 and uses thereof
US20220289866A1 (en)*2019-07-122022-09-15The Regents Of The University Of CaliforniaChemically controlled monoclonal antibody target engagement
WO2021207433A2 (en)*2020-04-072021-10-14The Regents Of The University Of CaliforniaEpitopes of sars-cov-2 neutralizing antibodies

Similar Documents

PublicationPublication DateTitle
Paduch et al.Generating conformation-specific synthetic antibodies to trap proteins in selected functional states
JP5675597B2 (en) Detection of cannabis use
US9146241B2 (en)Proteomic identification of antibodies
Harvey et al.An in silico method to assess antibody fragment polyreactivity
Lad et al.High-throughput kinetic screening of hybridomas to identify high-affinity antibodies using bio-layer interferometry
Ylera et al.Off-rate screening for selection of high-affinity anti-drug antibodies
Siegel et al.High efficiency recovery and epitope-specific sorting of an scFv yeast display library
KR102422494B1 (en)Method
Dyson et al.Mapping protein interactions by combining antibody affinity maturation and mass spectrometry
AU2015207665B2 (en)Cell surface prostate cancer antigen for diagnosis
Ayriss et al.High-throughput screening of single-chain antibodies using multiplexed flow cytometry
Conroy et al.Cardiac troponin I: a case study in rational antibody design for human diagnostics
Bharadwaj et al.Afucosylation of HLA-specific IgG1 as a potential predictor of antibody pathogenicity in kidney transplantation
Chung et al.Rare antibody phage isolation and discrimination (RAPID) biopanning enables identification of high-affinity antibodies against challenging targets
Kiguchi et al.Clonal array profiling of scFv-displaying phages for high-throughput discovery of affinity-matured antibody mutants
WO2024051096A1 (en)Anti-cd40 nanobody, and preparation method therefor and use thereof
Harth et al.Generation by phage display and characterization of drug-target complex-specific antibodies for pharmacokinetic analysis of biotherapeutics
Geuijen et al.Affinity ranking of antibodies using flow cytometry: application in antibody phage display-based target discovery
Koenig et al.Tuning the specificity of a Two-in-One Fab against three angiogenic antigens by fully utilizing the information of deep mutational scanning
Della Ducata et al.Solution equilibrium titration for high-throughput affinity estimation of unpurified antibodies and antibody fragments
WO2024249362A1 (en)Rare antibody phage isolation and discrimination (rapid) biological panning
Fournier et al.A platform for the early selection of non-competitive antibody-fragments from yeast surface display libraries
US20230304190A1 (en)Methods of screening antigen-binding molecules by normalizing for the concentration of antigen-binding molecule
Ferrara et al.Specific binder for Lightning-Link® biotinylated proteins from an antibody phage library
Hall et al.Quantitative specificity‐based display library screening identifies determinants of antibody‐epitope binding specificity

Legal Events

DateCodeTitleDescription
121Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number:24816245

Country of ref document:EP

Kind code of ref document:A1


[8]ページ先頭

©2009-2025 Movatter.jp