Methods of designing novel antibody mimetics for use in detecting antigens and as therapeutic agents

Information

  • Patent Grant
  • 11830582
  • Patent Number
    11,830,582
  • Date Filed
    Friday, June 14, 2019
    4 years ago
  • Date Issued
    Tuesday, November 28, 2023
    5 months ago
Abstract
Provided herein are methods implemented by a processor in a computer for designing a clamp peptide comprising the structure A1-B-A2 wherein A1 is a first peptide arm that binds to a first binding site of a target protein, A2 is a second peptide arm that binds to a second binding site of the target protein, and B is a bridge peptide which links A1 to A2. Also provided herein are computer-readable storage media having stored thereon machine-readable instructions executable by a processor and systems. Related methods of manufacturing a clamp peptide and the clamp peptides manufactures by the methods are provided.
Description
INCORPORATION BY REFERENCE OF MATERIAL SUBMITTED ELECTRONICALLY

Incorporated by reference in its entirety is a computer-readable nucleotide/amino acid sequence listing submitted concurrently herewith and identified as follows: 68,786 byte ASCII (Text) file named “53192A_Seqlisting.txt”; created on Jun. 12, 2019.


BACKGROUND

Antibodies constitute powerful therapeutic agents characterized by limited side effects due to their ability to specifically target a distinct antigen on a cell, bacteria, virus, or toxin. In 1986, the first therapeutic monoclonal antibody, Orthoclone OKT3, was introduced into the market. Since then, this class of biopharmaceutical products has significantly grown and over 45 monoclonal antibody products had received approval in the U.S. or Europe for the treatment of a variety of diseases, including cancer and inflammatory, cardiovascular, respiratory, and infectious diseases. Though the projected antibody market in the United States is anticipated to surpass the $10 billion mark, the production of such therapeutics is not without limitations. One disadvantage of therapeutic antibodies is the cost of downstream processing to achieve the required high purity levels. Another limiting factor of therapeutic antibodies is the sensitivity of antibody structure to chemical and physical denaturation encountered during post-manufacture filling, shipping and storage. Harsh chemical environments can affect the antibodies' binding properties.


Antibody mimetics have been successfully used in the development of binding assays for the detection of analytes in biological samples, as well as in separation methods, cancer therapy, targeted drug delivery, and in vivo imaging. The recent advances in the field of antibody mimetics and their applications in bioanalytical chemistry, specifically in diagnostics and other analytical methods have been described (Yu et al., Annual Review of Analytical Chemistry 10, 293-320 (2017). Because synthetic peptides are more resistant to physicochemical stress, characteristically more reproducible, and, ultimately, less expensive to manufacture and commercialize, when compared to antibodies, their use as elements of antibody mimetics capable of binding to ligand analytes in a manner analogous to that of the antigen-antibody interaction has spurred increased interest in the biotechnology and bioanalytical communities. To produce antibody mimetics that outperform antibodies with regard to binding affinities, cellular and tumor penetration, large-scale production, and temperature and pH stability is a high priority goal of the industry (Yu et al., 2017, supra).


In recent works, short peptides were used as molecular binders for virus detection. Linear peptides were selected by phage display to detect norovirus using an ELISA protocol or by means of an impedance biosensor (Hwang et al., 2017; Palzkill et al., 2018). Computationally-designed peptides were used to possibly detect flavivirus. The binding affinity and stability of disulfide cyclic peptide ligands with target Dengue virus (DENV) E glycoprotein were calculated by molecular docking and molecular dynamics simulation, but no experimental evidence was provided (Tambunan et al., 2016). A recent report predicted by molecular docking the structure of short peptides targeting the Zika virus (ZIKV) envelope protein and the interactions between the selected peptides and virus were assessed via a fluorescence-linked sandwich immunosorbent assay (FLISA), and the performance of the peptide-linked sandwich FLISA was evaluated in virus-spiked human serum and urine (Do Thi Hoang Kim et al., 2018).


While in silico methods of molecular modeling has been used to minimize experimental problems and facilitate the rational design of experimental protocols (Acebes et al., 2016; Bunker et al., 2016; Singh et al., 2016; Michaeli et al., 2018; Xu et al., 2018), and even though virtual docking is currently an important tool in drug discovery, and a subject of important developments over the last decade (Macalino et al., 2015; Yuan et al., 2017), a number of obstacles still limits the widespread use of molecular modeling for biotechnological applications. One of the most important drawbacks for mainstream use of molecular modeling is the challenge to simulate a huge number of candidates to be designed or/and docked using a full combinatorial approach.


Thus, improved methods of designing peptides for use in antibody mimetics is needed.


SUMMARY

Presented herein for the first time is a new methodology, based on an incremental construction approach, for the design and selection of short peptides that function as binding agents capable of selectively detecting target molecules. Herein, an in silico semi-combinatorial peptide screening method for designing the target-binding portions, the arms, of a new class of antibody mimetics, called clamp peptides, is demonstrated. In this virtual peptide screening method, different docking cycles of peptide libraries were generated and evaluated for binding to a binding site of a target protein. Using this method, the recognition properties of the amino acid motif between target binding and non-specific binding were maximized and allowed for ˜3 million peptides to be tested in a short period of time. Using this virtual screening method, clamp peptides designed to bind to two different binding sites on the ZIKV envelope protein were designed and subsequently manufactured and tested. As shown through direct ELISA, the performances of the clamp peptides demonstrated beneficial and desired binding activities. As shown by the data presented herein, the arms of the clamp peptides were able to wrap around the glycosylation site such as to clamp the peptide in place.


The present disclosure provides a method implemented by a processor in a computer for designing a clamp peptide comprising the structure A1-B-A2 wherein A1 is a first peptide arm that binds to a first binding site of a target protein, A2 is a second peptide arm that binds to a second binding site of the target protein, and B is a bridge peptide which links A1 to A2. In exemplary embodiments, the method comprises the steps of: (a) determining a binding score of each tetrapeptide of a first peptide library for the first binding site of the target protein, wherein the first peptide library comprises a set of tetrapeptides having a unique combination of four amino acids of the twenty naturally-occurring amino acids, optionally, wherein the first peptide library comprises the full combinatorial set of 160,000 tetrapeptides having a unique combination of four amino acids of the twenty naturally-occurring amino acids, (b) identifying the sequences of the tetrapeptides having a binding score which meets a first threshold, (c) determining a binding score of each pentapeptide of a second peptide library for the first binding site of the target protein, wherein the second peptide library comprises a set of pentapeptides having a unique combination of five amino acids comprising the amino acids of the sequence of a tetrapeptide identified in step (b) and one of the twenty naturally-occurring amino acids added as the first amino acid or the fifth amino acid of the pentapeptide or between two amino acids of the sequence of the tetrapeptide, and (d) identifying the sequences of the pentapeptides having a binding score which meets a second threshold. In exemplary embodiments, the method further comprises the steps of: (e) determining a binding score of each hexapeptide of a third peptide library for the first binding site of the target protein, wherein the third peptide library comprises a set of hexapeptides having a unique combination of six amino acids comprising the amino acids of the sequence of a pentapeptide identified in step (d) and one of the twenty naturally-occurring amino acids added as the first amino acid or the sixth amino acid of the hexapeptide or between two amino acids of the sequence of the pentapeptide and (f) identifying the sequences of the hexapeptides having a binding score which meets a third threshold. In exemplary embodiments, the method further comprises the steps of: repeating at least steps (a) to (d) wherein, for each determining step, a binding score for the second binding site of the target protein is determined for each tetrapeptide of the first peptide library and for each pentapeptide of the second peptide library. Optionally, wherein, when the method further comprises determining a binding score of each hexapeptide of a third peptide library for the first binding site of the target protein, the method further comprises repeating these steps, wherein for each determining step, a binding score for the second binding site of the target protein is determined for each hexapeptide of the third peptide library.


The present disclosure also provides a computer-readable storage medium having stored thereon machine-readable instructions executable by a processor, comprising instructions for carrying out the steps of any one of the presently disclosed methods for designing a clamp peptide.


Further provided is a system comprising machine readable instructions that, when executed by the processor, cause the processor to carry out the steps of any one of the presently disclosed methods for designing a clamp peptide.


The present disclosure also provides a method of manufacturing a clamp peptide comprising the structure A1-B-A2 wherein A1 is a first peptide arm that binds to a first binding site of a target protein, A2 is a second peptide arm that binds to a second binding site of the target protein, and B is a bridge peptide which links A1 to A2. In exemplary embodiments, the method comprises (i) designing the first peptide arm and the second peptide arm of the clamp peptide according to any one of the presently disclosed methods for designing a clamp peptide and (ii) joining the first peptide arm to the second peptide arm with a bridge peptide, B.


A clamp peptide comprising the structure A1-B-A2 wherein A1 is a first peptide arm that binds to a first binding site of a target protein, A2 is a second peptide arm that binds to a second binding site of the target protein, and B is a bridge peptide which links A1 to A2, wherein the sequences of A1 and A2 are designed by any one of the presently disclosed methods for designing a clamp peptide.


Use of the presently disclosed clamp peptides for detecting target proteins, e.g., antigens, are furthermore provided herein. Also, use of the presently disclosed clamp peptides for treating a subject in need thereof are provided herein.


Additional descriptions and guidance, as well as exemplification, of the presently disclosed methods are provided herein.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1A is a computer-generated illustration of the target protein (ZIVA envelope protein) and the two active sites chosen for docking the peptide libraries. The larger blue shapes represent the electrostatic surfaces of the active sites. FIG. 1B is a schematic representation of the clamp peptide structure made by two peptide arms of five or six amino acids in length linked by a bridge peptide. FIG. 1C is a computer-generated illustration of the target protein of FIG. 1A with two peptides (outlined in orange) docked in the two active sites. The larger blue shaded regions represent the electrostatic surfaces of the active sites. The length (in angstrom) between the two docked peptides is shown (19.31 Å or 1.931 nm) in yellow. FIG. 1D is a computer-generated illustration of a bridge peptide designed and minimized in zwitterionic mode. The bridge peptide comprises eight amino acids long and has the amino acid sequence GPGCCGPG (SEQ ID NO: 32). The length in angstrom is shown (19.56 Å=1.956 nm).



FIG. 2A is a graph of the binding score (kcal/mol) of the three peptides libraries (first library of tetrapeptides (blue); second peptide library of pentapeptides (orange); third peptide library of hexapeptides (gray)) docked in the active site 1 (Asn154), showing the typical distribution of scores obtained in the simulations. The binding score data were sorted in ascending order of binding score, thus not necessarily a correspondence must exist between the positions of the peptides in each curve. FIG. 2B is a schematic representation of the semi-combinatorial approach for obtaining penta- and hexapeptide libraries. The semi-combinatorial approach is based on generating different cycles of peptide libraries by maximizing the recognition properties of amino acid motif between the ZIKV binding site and the other flaviviruses binding sites.



FIG. 3 is a series of graphs depicting the sigmoidal ZIKV particles concentration response trend. Y axis=A absorbance (450 nm); X-axis=log [ZIKV], copies/mL.



FIG. 4 is a graph of the A spectrophotometric absorbance signals obtained in a cross-reactivity study using the ELISA direct assay for the best three clamp peptides (C3, C6 and C7) and two mono-arm peptides (P2 and X1) binding the ZIKV target protein (yellow) and three serotypes of DENV virus (DENV-1 (blue), DENV-2 (orange) and DENV-3 (grey)) at the concentration of 105 [ZIKV] copies/mL.



FIG. 5 is a graph of the A spectrophotometric absorbance signals obtained in a study using the ELISA direct assay for the best three clamp peptides (C3, C6 and C7) and two mono-arm peptides (P2 and X1) binding the ZIKV target protein at a concentration of 105 copies/ml (blue) or 106 copies/mL (orange) in the presence of buffer (*), serum (s), or urine (u).



FIG. 6A is a schematic of an exemplary embodiment 101 of a system 100 for designing a clamp peptide. FIG. 6B is a schematic of base routines that may be stored on non-volatile memory of the system 100 of FIG. 6A. FIG. 6C is a schematic of exemplary sub-routines for the base routines of FIG. 6B.





DETAILED DESCRIPTION

The present disclosure provides a method implemented by a processor in a computer for designing a clamp peptide. Clamp peptides, as further described herein, represent a new class of antibody mimetics. In exemplary aspects, the clamp peptide comprises the structure A1-B-A2 wherein A1 is a first peptide arm that binds to a first binding site of a target protein, A2 is a second peptide arm that binds to a second binding site of the target protein, and B is a bridge peptide which links A1 to A2. In exemplary aspects, each of A1 and A2 is a peptide of about 5 to about 7 amino acids, e.g., about 5 amino acids, about 6 amino acids, or about 7 amino acids. In exemplary aspects, the bridge peptide (B) is about 5 to about 10 amino acids in length (e.g., about 5 amino acids, about 6 amino acids, about 7 amino acids, about 8 amino acids, about 9 amino acids, or about 10 amino acids in length. In various aspects, the amino acids of the clamp peptide, or the first peptide arm, the second peptide arm, or the bridge peptide thereof, are naturally-occurring or coded or non-naturally occurring or non-coded. Non-naturally occurring amino acids refer to amino acids that do not naturally occur in vivo but which, nevertheless, can be incorporated into the peptide structures described herein. “Non-coded” as used herein refer to an amino acid that is not an L-isomer of any of the following 20 amino acids: Ala, Cys, Asp, Glu, Phe, Gly, His, Ile, Lys, Leu, Met, Asn, Pro, Gln, Arg, Ser, Thr, Val, Trp, Tyr. Naturally-occurring or coded amino acids include the L-isomers of the amino acids: Ala, Cys, Asp, Glu, Phe, Gly, His, Ile, Lys, Leu, Met, Asn, Pro, Gln, Arg, Ser, Thr, Val, Trp, Tyr. Exemplary non-naturally occurring or non-coded amino acids include but are not limited to, any D or L isomers of the non-naturally occurring or non-coded amino acids selected from the group consisting of: beta-alanine, N-alpha-methyl-alanine (Me-Ala), aminobutyric acid (Abu), gamma-aminobutyric acid, aminohexanoic acid (epsilon-Ahx), aminoisobutyric acid (Aib), aminomethylpyrrole carboxylic acid, aminopiperidinecarboxylic acid, aminoserine (Ams), am inotetrahydropyran-4-carboxylic acid, arginine N-methoxy-N-methyl amide, beta-aspartic acid (beta-Asp), azetidine carboxylic acid, 3-(2-benzothiazolyl)alanine, alpha-tert-butylglycine, 2-amino-5-ureido-n-valeric acid (citrulline, Cit), beta-Cyclohexylalanine (Cha), acetamidomethyl-cysteine, diaminobutanoic acid (Dab), diaminopropionic acid (Dpr), dihydroxyphenylalanine (DOPA), dimethylthiazolidine (DMTA), gamma-Glutamic acid (gamma-Glu), homoserine (Hse), hydroxyproline (Hyp), isoleucine N-methoxy-N-methyl amide, methyl-isoleucine (Melle), isonipecotic acid (lsn), methyl-leucine (MeLeu), methyl-lysine, dimethyl-lysine, trimethyl-lysine, methanoproline, methionine-sulfoxide (Met(O)), methionine-sulfone (Met(O2)), norleucine (Nle), methyl-norleucine (Me-Nle), norvaline (Nva), ornithine (Orn), para-aminobenzoic acid (PABA), penicillamine (Pen), methylphenylalanine (MePhe), 4-Chlorophenylalanine (Phe(4-Cl)), 4-fluorophenylalanine (Phe(4-F)), 4-nitrophenylalanine (Phe(4-NO2)), 4-cyanophenylalanine ((Phe(4-CN)), phenylglycine (Phg), piperidinylalanine, piperidinylglycine, 3,4-dehydroproline, pyrrolidinylalanine, sarcosine (Sar), selenocysteine (Sec), O-Benzyl-phosphoserine, 4-amino-3-hydroxy-6-methylheptanoic acid (Sta), 4-amino-5-cyclohexyl-3-hydroxypentanoic acid (ACHPA), 4-amino-3-hydroxy-5-phenylpentanoic acid (AHPPA), 1,2,3,4,-tetrahydro-isoquinoline-3-carboxylic acid (Tic), tetrahydropyranglycine, thienylalanine (Thi), O-benzyl-phosphotyrosine, O-Phosphotyrosine, methoxytyrosine, ethoxytyrosine, O-(bis-dimethylamino-phosphono)-tyrosine, tyrosine sulfate tetrabutylamine, methyl-valine (MeVal), and alkylated 3-mercaptopropionic acid. In some embodiments, the clamp peptide, or the first peptide arm, the second peptide arm, or the bridge peptide thereof, described herein are glycosylated, amidated, carboxylated, phosphorylated, esterified, N-acylated, cyclized via, e.g., a disulfide bridge, or converted into a salt (e.g., an acid addition salt, a basic addition salt), and/or optionally dimerized, multimerized, or polymerized, or conjugated.


In exemplary embodiments, the method implemented by a processor in a computer for designing a clamp peptide comprises the steps of: (a) determining a binding score of each tetrapeptide of a first peptide library for the first binding site of the target protein, wherein the first peptide library comprises a set of tetrapeptides having a unique combination of four amino acids of the twenty naturally-occurring amino acids, optionally, wherein the first peptide library comprises the full combinatorial set of 160,000 (160K) tetrapeptides having a unique combination of four amino acids of the twenty naturally-occurring amino acids, (b) identifying the sequences of the tetrapeptides having a binding score which meets a first threshold, (c) determining a binding score of each pentapeptide of a second peptide library for the first binding site of the target protein, wherein the second peptide library comprises a set of pentapeptides having a unique combination of five amino acids comprising the amino acids of the sequence of a tetrapeptide identified in step (b) and one of the twenty naturally-occurring amino acids added as the first amino acid or the fifth amino acid of the pentapeptide or between two amino acids of the sequence of the tetrapeptide, and (d) identifying the sequences of the pentapeptides having a binding score which meets a second threshold.


In exemplary embodiments, the method further comprises the steps of: (e) determining a binding score of each hexapeptide of a third peptide library for the first binding site of the target protein, wherein the third peptide library comprises a set of hexapeptides having a unique combination of six amino acids comprising the amino acids of the sequence of a pentapeptide identified in step (d) and one of the twenty naturally-occurring amino acids added as the first amino acid or the sixth amino acid of the hexapeptide or between two amino acids of the sequence of the pentapeptide and (f) identifying the sequences of the hexapeptides having a binding score which meets a third threshold. In exemplary embodiments, the method further comprises the steps of: repeating at least steps (a) to (d) wherein, for each determining step, a binding score for the second binding site of the target protein is determined for each tetrapeptide of the first peptide library and for each pentapeptide of the second peptide library. Optionally, wherein, when the method further comprises determining a binding score of each hexapeptide of a third peptide library for the first binding site of the target protein, the method further comprises repeating these steps, wherein for each determining step, a binding score for the second binding site of the target protein is determined for each hexapeptide of the third peptide library.


In exemplary embodiments, the method further comprises (I) determining a binding score of each heptapeptide of a fourth peptide library for the first binding site of the target protein, wherein the fourth peptide library comprises a set of heptapeptides having a unique combination of seven amino acids comprising the amino acids of the sequence of a hexapeptide identified in step (f) and one of the twenty naturally-occurring amino acids added as the first amino acid or the seventh amino acid of the hexapeptide or between two amino acids of the sequence of the hexapeptide and (II) identifying the sequences of the heptapeptides having a binding score which meets a fourth threshold. In exemplary embodiments, the method further comprises the steps of: repeating at least steps of the method, wherein, for each determining step, a binding score for the second binding site of the target protein is determined for each tetrapeptide of the first peptide library and for each pentapeptide of the second peptide library. Optionally, wherein, when the method further comprises determining a binding score of each hexapeptide of a third peptide library and a binding score of each heptapeptide for the first binding site of the target protein, the method further comprises repeating these steps, wherein for each determining step, a binding score for the second binding site of the target protein is determined for each hexapeptide of the third peptide library and for each heptapeptide of the fourth peptide library.


In exemplary embodiments, the method does not further comprise determining a binding score of each heptapeptide of a fourth peptide library. In exemplary instances, the third peptide library is the last peptide library for which binding scores are determined.


The method in some aspects comprises generating the first peptide library. Optionally, the first peptide library comprises 160,000 unique tetrapeptides, optionally, the full combinatorial set of 160,000 tetrapeptides having a unique combination of four amino acids of the twenty naturally-occurring amino acids.


The method in some aspects comprises generating the second peptide library. In various instances, the second peptide library comprises pentapeptides having a sequence based on the sequences of the tetrapeptides (post-application of the first threshold) and another amino acid. In various aspects, the second peptide library comprises a set of pentapeptides having a unique combination of five amino acids comprising the amino acids of the sequence of a tetrapeptide identified in step (b) and one of the twenty naturally-occurring amino acids added as the first amino acid or the fifth amino acid of the pentapeptide or between two amino acids of the sequence of the tetrapeptide.


The method in some aspects comprises generating the third peptide library. In various instances, the third peptide library comprises hexapeptides having a sequence based on the sequences of the pentapeptides (post-application of the second threshold) and another amino acid. In various aspects, the third peptide library comprises a set of hexapeptides having a unique combination of six amino acids comprising the amino acids of the sequence of a pentapeptide identified in step (d) and one of the twenty naturally-occurring amino acids added as the first amino acid or the sixth amino acid of the hexapeptide or between two amino acids of the sequence of the pentapeptide.


In various instances, the first threshold is a binding score within the top 5% of binding scores (e.g., top binding scores mean the strongest binding between peptide and active site of target protein). In various aspects, approximately 8000 unique sequences of tetrapeptides are identified upon application of the first threshold. In certain aspects, the first threshold is a binding score within the top 5% of binding scores and a binding score outside the top 5% of binding scores for a different target protein. In some instances, about 1000 unique sequences of tetrapeptides are upon application of the first threshold. In exemplary instances, the second threshold is the same as the first threshold. In alternative instances, the first threshold is different from the second threshold. In exemplary instances, the third threshold is the same as the first threshold or the second threshold. In alternative instances, the third threshold is different from the second threshold and/or first threshold. In various aspects, the third threshold is a binding score within the top 5% of binding scores. In certain instances, the third threshold further comprises a binding score outside the top 5% of binding scores for a different target protein.


The binding scores are determined using a molecular docking program, optionally, a molecular docking program that is based on multi-conformer rigid body docking, which evaluates several conformers per peptide. The binding scores in some aspects are calculated using a docking scoring function. In some aspects, one or more of LUDI, Chemscore, chemgauss4, DOCK, FlexX, Gold, Pmf, Score, Fresno, AutoDock Vina, Dynadock, LigScore, Rosetta FlexPepDock are used for determining and/or calculating the binding score. Such docking scoring functions are known in the art. See, e.g., Logean et al., Bioorganic and Medicinal Chem Lett 11(5): 675-679 (2001), Trott et al., J Computational Chem https://doi.org/10.1002/jcc.21334; Bohm et al., J Computer-Aided Molecular Design 13(1): 51-56 (1999); Raveh et al., PLoS ONE 6(4): e18934. https://doi.org/10.1371/journal.pone.0018934; Krammer et al., J Molecular Graphics and Modelling 23(5): 395-407 (2005). In various instances, the binding scores for more than 350,000 peptides are determined for each of the first binding site and the second binding site.


The method in certain instances further comprises determining the length of B of the clamp peptide, optionally, by measuring the distance between a peptide bound to the first binding site of the target protein and a peptide bound to the second binding site of the target protein. The bridge peptide of the clamp peptide in various aspects is designed based on the length determined.


The present disclosure also provides a computer-readable storage medium having stored thereon machine-readable instructions executable by a processor, comprising instructions for carrying out the steps of any one of the presently disclosed methods for designing a clamp peptide.


Further provided is a system comprising machine readable instructions that, when executed by the processor, cause the processor to carry out the steps of any one of the presently disclosed methods for designing a clamp peptide.



FIG. 6A illustrates an exemplary embodiment 101 of a system 100 for designing a clamp peptide. Generally, the system 100 may include one or more client devices 102, a network 104, and/or a network-accessible database 108. Each client device 102 may be communicatively coupled to the network 104 by one or more wired or wireless network connections 112, which may be, for example, a connection complying with a standard such as one of the IEEE 802.11 standards (“Wi-Fi”), the Ethernet standard, or any other appropriate network connection. Similarly, the database 108 may be communicatively coupled to the network 104 via one or more connections 114. (Of course, the database could alternatively be internal to one or more of the client devices 102.) The database 108 may store the sequences of each peptide of a peptide library, optionally, the sequences of each tetrapeptide of the first peptide library, the sequences of each pentapeptide of the second peptide library, the sequences of each hexapeptide of the third peptide library. The database 108 additionally or alternatively may store the binding score of each peptide for each peptide library, and/or the identity of the peptide(s) which meet the threshold, and/or the thresholds themselves.


As will be understood, the network 104 may be a local area network (LAN) or a wide-area network (WAN). That is, network 104 may include only local (e.g., intra-organization) connections or, alternatively, the network 104 may include connections extending beyond the organization and onto one or more public networks (e.g., the Internet). In some embodiments, for example, the client device 102 and the database 108 may be within the network operated by a single company (Company A). In other embodiments, for example, the client device(s) 102 may be on a network operated by Company A, while the database 108 may be on a network operated by a second company (Company B), and the networks of Company A and Company B may be coupled by a third network such as, for example, the Internet.


Referring still to FIG. 6A, the client device 102 includes a processor 128 (CPU), a RAM 130, and a non-volatile memory 132. The non-volatile memory 132 may be any appropriate memory device including, by way of example and not limitation, a magnetic disk (e.g., a hard disk drive), a solid state drive (e.g., a flash memory), etc. Additionally, it will be understood that, at least with regard to FIG. 6A, the database 108 need not be separate from the client device 102. Instead, in some embodiments, the database 108 is part of the non-volatile memory 132 and the data 122, 124, 126 may be stored as data within the memory 132. The database may comprise, for instance, the sequences of each tetrapeptide of the first peptide library. The database may further comprise claim data 122 which comprises, for example, the binding scores for each of the tetrapeptides of the first peptide library. Likewise, the database may comprise the sequences of each pentapeptide of the second peptide library and the binding scores for each of the pentapeptides of the second library. Also, the database in some aspects comprises the sequences of each hexapeptide of the third peptide library and the binding scores for each of the hexapeptides of the third library. The benchmark data 124 may comprise binding scores for commercial antibodies to the target protein, which may serve as a benchmark against which the binding scores of the peptides of the peptide libraries may be compared.


For example, the data 122 may be included as data in a spreadsheet file stored in the memory 132, instead of as data in the database 108. In addition to storing the records of the database 108 (in some embodiments), the memory 132 stores program data and other data necessary to analyze data (e.g., binding scores) of one or more peptide libraries, determine binding scores, identify the sequences of the peptides which meet a threshold, the thresholds themselves. For example, in an embodiment, the memory 132 stores a first routine 134, a second routine 136, and a third routine 138. The first routine 134 may determine binding scores of each tetrapeptide of a first peptide library for a binding site of the target protein. The second routine 136 may compute one or more statistical parameters of the binding scores collected by the first routine 134, and/or apply a threshold to the binding scores to identify the tetrapeptides (e.g., identify the sequences of the tetrapeptides) meeting that threshold. The third routine 138 may, for example, generate the sequences of pentapeptides of the second peptide library based on the sequences of the tetrapeptides that met the threshold, as identified by the second routine. In exemplary embodiments, the memory 132 stores a first routine 134, a second routine 136, and a third routine 138, as outlined above, and additionally stores a fourth routine, a fifth routine, and a sixth routine. The fourth routine may determine binding scores of each pentapeptide of the second peptide library generated by the third routine for a binding site of the target protein. The fifth routine may compute one or more statistical parameters of the binding scores collected by the fourth routine 134, and/or apply a threshold to the binding scores to identify the pentapeptides (e.g., identify the sequences of the pentapeptides) meeting that threshold. The sixth routine may generate the sequences of hexapeptides of the third peptide library based on the sequences of the pentapeptides that met the threshold, as identified by the fifth routine. The memory 132 may store additional routines for subsequent cycles of determining binding scores of peptides of a peptide library, applying a threshold to identify peptides that meet the threshold, and generating the sequences of the peptides of the next peptide library using the sequences of the peptides meeting the threshold (as identified by the immediately prior routine). For instance, the memory 132 may store a seventh routine, eighth routine, and ninth routine, wherein the seventh routine may determine binding scores of each hexapeptide of a third peptide library for a binding site of the target protein, the eighth routine may compute one or more statistical parameters of the binding scores collected by the seventh routine, and/or apply a threshold to the binding scores to identify the hexapeptides (e.g., identify the sequences of the hexapeptides) meeting that threshold. In exemplary aspects, the memory 132 may store the routines described in FIGS. 6B and 6C.


Regardless, each of the routines is executable by the processor 128 and comprises a series of compiled or compilable machine-readable instructions stored in the memory 132. Additionally, the memory 132 may store generated reports or records of data output by one of the routines 134 or 136. Alternatively, the reports or records may be output to the database 108. One or more display/output devices 140 (e.g., printer, display, etc.) and one or more input devices 142 (e.g., mouse, keyboard, tablet, touch-sensitive interface, etc.) may also be coupled to the client device 102, as is generally known.


As will be understood, although individual operations of one or more methods are illustrated and described as separate operations, one or more of the individual operations may be performed concurrently, and nothing requires that the operations be performed in the order illustrated. Structures and functionality presented as separate components in example configurations may be implemented as a combined structure or component. Similarly, structures and functionality presented as a single component may be implemented as separate components. These and other variations, modifications, additions, and improvements fall within the scope of the subject matter herein.


For example, the network 104 may include but is not limited to any combination of a LAN, a MAN, a WAN, a mobile, a wired or wireless network, a private network, or a virtual private network. Moreover, while only two clients 102 are illustrated in FIG. 6A to simplify and clarify the description, it is understood that any number of client computers are supported and can be in communication with one or more servers (not shown).


Additionally, certain embodiments are described herein as including logic or a number of routines. Routines may constitute either software routines (e.g., code embodied on a machine-readable medium or in a transmission signal) or hardware routines. A hardware routine is tangible unit capable of performing certain operations and may be configured or arranged in a certain manner. In example embodiments, one or more computer systems (e.g., a standalone, client or server computer system) or one or more hardware routines of a computer system (e.g., a processor or a group of processors) may be configured by software (e.g., an application or application portion) as a hardware routine that operates to perform certain operations as described herein.


Similarly, the methods or routines described herein may be at least partially processor-implemented. For example, at least some of the operations of a method may be performed by one or processors or processor-implemented hardware modules. The performance of certain of the operations may be distributed among the one or more processors, not only residing within a single machine, but deployed across a number of machines. In some example embodiments, the processor or processors may be located in a single location (e.g., within a home environment, an office environment or as a server farm), while in other embodiments the processors may be distributed across a number of locations.


The performance of certain of the operations may be distributed among the one or more processors, not only residing within a single machine, but deployed across a number of machines. In some example embodiments, the one or more processors or processor-implemented modules may be located in a single geographic location (e.g., within a home environment, an office environment, or a server farm). In other example embodiments, the one or more processors or processor-implemented modules may be distributed across a number of geographic locations.


Some embodiments may be described using the expression “coupled” and “connected” along with their derivatives. For example, some embodiments may be described using the term “coupled” to indicate that two or more elements are in direct physical or electrical contact. The term “coupled,” however, may also mean that two or more elements are not in direct contact with each other, but yet still co-operate or interact with each other. The embodiments are not limited in this context.


As used herein, the terms “comprises,” “comprising,” “includes,” “including,” “has,” “having” or any other variation thereof, are intended to cover a non-exclusive inclusion. For example, a process, method, article, or apparatus that comprises a list of elements is not necessarily limited to only those elements but may include other elements not expressly listed or inherent to such process, method, article, or apparatus. Further, unless expressly stated to the contrary, “or” refers to an inclusive or and not to an exclusive or. For example, a condition A or B is satisfied by any one of the following: A is true (or present) and B is false (or not present), A is false (or not present) and B is true (or present), and both A and B are true (or present).


In addition, use of the “a” or “an” are employed to describe elements and components of the embodiments herein. This is done merely for convenience and to give a general sense of the description. This description should be read to include one or at least one and the singular also includes the plural unless it is obvious that it is meant otherwise.


Still further, the figures depict preferred embodiments of a map editor system for purposes of illustration only. One skilled in the art will readily recognize from the following discussion that alternative embodiments of the structures and methods illustrated herein may be employed without departing from the principles described herein


Upon reading this disclosure, those of skill in the art will appreciate still additional alternative structural and functional designs for a system and a process for identifying terminal road segments through the disclosed principles herein. Thus, while particular embodiments and applications have been illustrated and described, it is to be understood that the disclosed embodiments are not limited to the precise construction and components disclosed herein. Various modifications, changes and variations, which will be apparent to those skilled in the art, may be made in the arrangement, operation and details of the method and apparatus disclosed herein without departing from the spirit and scope defined in the appended claims.


Methods of manufacturing a clamp peptide are further provided herein. In various aspects, the clamp peptide comprises the structure A1-B-A2 wherein A1 is a first peptide arm that binds to a first binding site of a target protein, A2 is a second peptide arm that binds to a second binding site of the target protein, and B is a bridge peptide which links A1 to A2. In exemplary embodiments, the method comprises (I) designing the first peptide arm and the second peptide arm of the clamp peptide according to any one of the presently disclosed methods implemented by a processor in a computer for designing a clamp peptide and (II) joining the first peptide arm to the second peptide arm with a bridge peptide, B. In exemplary aspects, the method further comprises assaying the binding of the clamp peptide to the target protein. Suitable methods of assaying peptide-protein binding are known in the art and include for instance assays based on Western blotting, an enzyme-linked immunosorbent assay (ELISA), a radioimmunoassay (RIA), and immunohistochemical assay, and ligand binding assays (LBAs) which are described in O'Hara et al., AAPS J 14(2): 316-328 (2012).


The clamp peptides manufactured by any one of the presently disclosed methods of manufacturing a clamp peptide are further provided herein.


The following examples are given merely to illustrate the present invention and not in any way to limit its scope.


EXAMPLES
Example 1

This example describes an exemplary method of designing clamp peptides, a new generation of antibody mimetics.


Abstract


A new class of antibody mimetics is proposed. This is a generic universal approach that can be used against any type of antigen. The antibody mimetics were designed connecting three short peptides mimicking a clamp with two arms and one bridge.


The peptide arms were computationally designed using a virtual approach based on generating different docking cycles of tetra, penta, hexapeptide libraries by maximizing the recognition properties of amino acid motifs between the ZIKV envelope protein active sites and other flaviviruses binding sites (DENV, Chikungunya and West Nile).


The peptide bridge, connecting the two arms, was made with glycine for spacing, proline for bending and two cysteines for biotin functionalization.


Eight clamp peptides and four mono arm peptides were then synthesized and tested vs intact ZIKV particles by using a direct enzyme linked immunosorbent assay (ELISA). As a reference, we employed a well-established anti-ZIKV virus antibody, the antibody 4G2.


Three clamp peptides assay showed a detection limit one or two order of magnitude lower (around 3.16*103 [ZIKV] copies/mL) then the antibody or mono-arm peptides with a dynamic range from 104 to 107 copies/mL of intact ZIKV particles. Synthetic clamp peptides showed low coefficient of variation (<5%) and a good inter-day and batch to batch reproducibility (<15%). These three clamp peptides showed slight cross-reactivity against three serotypes of DENV (DENV-1, -2 and -3) at the concentration of 10{circumflex over ( )}5 copies/mL of intact virus particles, but the discrimination between the DENV and ZIKV was lost increasing the coating concentration to 10{circumflex over ( )}6 copies/mL of the viruses.


The sensitivity of the clamp peptides was tested in the presence of two biological matrices, urine and serum diluted 1:1 and 1:10, respectively. The detection limits of clamp peptides decreased about one order of magnitude for ZIKV detection in urine or serum, with a distinct analytical signal starting from 10{circumflex over ( )}5 copies/mL of ZIKV.


Introduction


The threat of ZIKV infection has emerged as a global public health problem because of its ability to cause severe congenital disease and affect a large population (loos et al., 2014; Weaver et al., 2016). ZIKV infection is known to cause neurological problems to pregnant women and potentially cause microcephaly and other congenital malformations and diseases to the unborn child. ZIKV affects, both male and females and it has been reported that the virus can be transmitted sexually through semen and vaginal fluids. The ZIKV virus is a mosquito-borne flavivirus, and due to the lack of specific antibodies/binders that can be used for diagnosis of the disease, the current bioassays present cross-reactivity with other flaviviruses and arboviruses. It is well established that ZIKV has many common genetic sequences and protein structures with other flaviviruses, like DENV, West Nile virus or Chikungunya (Barba-Spaeth et al., 2016; Heffron et al., 2018). This limits the use of immunoassays for the detection of human pathogens within the flavivirus genus (Priyamvada et al., 2016; Stettler et al., 2016). Thus, there is a need for highly selective binders for ZIKV that can be employed in diagnostics and health status assessment of patients suffering from ZIKV.


The flavivirus envelope protein is responsible for virus entry and represents a major target for neutralizing antibodies. The ZIKV virus structure is similar to other known flaviviruses structures except for the ˜10 amino acids that surround the Asn-154 glycosylation site found in each of the 180 envelope glycoproteins that make up the icosahedral shell (Zhao et al., 2016).


In this work, the clamp peptide arms were designed using as guide the crystallographic coordinates of the ZIKV envelope protein glycosylation binding site. The entire molecular surface of the glycosylation envelope protein biding site was defined by two cubic boxes where arm peptides were expected to bind.


The in-silico screening technique was based on a semi-combinatorial approach by designing peptides that could wrap around the glycosylation site such as to clamp the peptide in place. Different docking cycles of peptide libraries were generated by maximizing the recognition properties of the amino acid motif between the ZKV glycosylation site and the other flaviviruses glycosylation binding sites (DENV, Chikungunya and West Nile). A total library of around three million peptides was tested in-silico.


Peptides as antibody mimetic elements in diagnostic methods were recently reviewed highlighting the features desired to outperform antibodies with regard to binding affinities, cellular and tumor penetration, large-scale production, temperature, and pH stability (Yu et al., 2017). It is well documented that a hard chemical environment can affect antibodies binding properties and DNA and peptides aptamers are the most promising candidates to replace them in bioanalysis as reported by recent reviews (Gong et al., 2010; Pichon et al., 2015; Li et al., 2018). Aptamers have become increasingly important molecular tools for diagnostics and as therapeutic agents, and are used in many analytical applications, such as chromatography, electrophoresis, mass spectrometry, molecular beacons, gas sensors, and biosensors (Stobiecka and Chalupa, 2015; Lin et al., 2016; Tang et al., 2016; Mascini et al., 2017; Mascini et al., 2019).


In recent works, short peptides were used as molecular binders for virus detection. Linear peptides were selected by phage display to detect norovirus using an ELISA protocol or by means of an impedance biosensor (Hwang et al., 2017; Palzkill et al., 2018).


Computationally designed peptides were used to possibly detect flavivirus. Binding affinity and stability of disulfide cyclic peptide ligands with target DENV E glycoprotein were calculated by molecular docking and molecular dynamics simulation, but no experimental evidence was provided (Tambunan et al., 2016).


A recent report predicted by molecular docking the structure of short peptides targeting the ZIKV envelope protein and the interactions between the selected peptides and virus were assessed via a fluorescence-linked sandwich immunosorbent assay (FLISA), and the performance of the peptide-linked sandwich FLISA was evaluated in virus-spiked human serum and urine (Do Thi Hoang Kim et al., 2018).


Molecular modelling is more and more used to overcome the trial and error approach and to minimize experimental problems by providing an understanding of atomic interactions and facilitating the rational design of experimental protocols (Acebes et al., 2016; Bunker et al., 2016; Singh et al., 2016; Michaeli et al., 2018; Xu et al., 2018). Virtual docking is currently an important tool in drug discovery, and a subject of important developments over the last decade (Macalino et al., 2015; Yuan et al., 2017).


However, a number of obstacles still limits the widespread use of molecular modelling for biotechnological applications. One of the most important drawbacks for mainstream use of molecular modeling is the challenge to simulate a huge number of candidates to be designed or/and docked using a full combinatorial approach.


To address this issue, herein we present a new methodology, based on an incremental construction approach to choose short peptides as binding agents for the selective detection of the intact ZIKV particles. Synthetic peptides are more resistant to physicochemical stress, more reproducible and less expensive when compared with antibodies so even if they show less specificity can be used as an array giving synergetic contribution to the detection.


Direct ELISA was chosen as the experimental protocol to check the performances of the clamp peptides. ELISA was preferred to other analytical techniques because it provides automated steps to speed-up the screening of a large number of experimental trials.


Materials and Methods


All calculations of molecular docking were done using a desktop computer with 19 processors Intel Xeon X5690 at 3.47 GHz each, with 94.5 GiB RAM, running Kernel Linux 2.6.32-642.1.1e16.x86_64, GNOME 2.28.2.


The three peptide libraries were designed and cleaned up with Hyperchem 8.0.5.


Peptides were designed in zwitterionic mode, using only the 20 natural amino acids, adding hydrogens, using molecular mechanics method amber, with the algorithm “Steepest Descents” converging at 0.08 Kj mol−1 in 32767 as maximum of cycles. Minimization, conformers generation and docking were carried out using Open Eye Scientific Software package under academic license. Each peptide library was compacted in a single file and fast minimized in gas phase to reduce computing time. In this context, solvent condition did not change significantly the results. The energy minimization process was carried out using SZYBKI 1.5.7 in its default parameterization (SZYBKI, version 1.5.7). To take into account the flexibility of the peptides, ten conformers were generated for each peptide by means of the OMEGA 2.4.6 used with MMFF as the force field (Hawkins et al., 2010; Hawkins and Nicholls, 2012; OMEGA, version 2.4.6). Therefore, the ligands were represented by the peptide conformers, around 5 millions units.


Then the envelope proteins, taken as the receptors, were downloaded from the protein data bank web site. The envelope proteins were from the flavivirus species ZIKV DENV, Chikungunya and West Nile having respectively the following codes in the Protein Data Bank web site: SIRE, 4UTC, 3N40 and 3I50. All residues and water molecules were removed from the envelope proteins pdb files. For each envelope protein two dedicated boxes were generated, one enfolding the glycosylation site the other in the closet cavity from the glycosylation site.


In order to reduce the calculation time, tetra and pentapeptide libraries were docked using active site boxes with a volume of around 13 nm3 and hexapeptide libraries using boxes having a volume of around 18 nm3.


Using these sizes, the entire molecular surface of all peptide conformers was inside the active site box.


The active site box along with the Multi-conformer rigid body docking were carried out using OEDocking 3.0.0 (Kelley et al., 2015; OEDocking, version 3.0.0). Multi-conformer rigid body docking was run using Chemgauss4 as scoring function. The Chemgauss4, a modification of the Chemgauss3, was the latest scoring function from OpenEye software with improved hydrogen bonding and metal chelator functions. The total score obtained was the sum of steric, acceptor/metal, donor and aromatic contributions. The time required for docking a peptide library was about 24 hours.


Structures visualization and generation of molecular surfaces were performed using VIDA 4.1.1 (VIDA, version 4.1.1).


The entire process was automated using a bash script and using a freeware BASIC-like scripting language (AutoIT V3) for post processing data analysis.


Experimental Setup


All chemicals used for buffers were of analytical grade and purchased from Sigma-Aldrich (http://www.sigmaaldrich.com).


The eight clamp peptides and the four mono-arm peptides were purchased from Biomatik (http://www.biomatik.com). Cysteines within the peptide structures were used to bind maleimide-PEG2-biotin. All peptides were provided with a purity >85%.


Lyophilized peptides were diluted at 1 mM concentration in 10 mM phosphate buffered saline (PBS) pH 7.4, divided into 1004 aliquots and stored at −30° C. for further use.


Before biotin functionalization, peptides stock solution was reduced using trialkylphosphine (TCEP) from ThermoFisher Scientific (www.thermofisher.com) and after 1 h the gel was removed using TCEP gel spin separation columns (ThermoFisher Scientific). Then, 2-fold molar excess of EZ-Link™ Maleimide-PEG2-Biotin (ThermoFisher Scientific) was added to purified peptide solution and incubated for 1 h. At this concentration EZ-Link™ Maleimide-PEG2-Biotin did not contribute to background signal as shown by a pilot test using only EZ-Link™ Maleimide-PEG2-Biotin without peptide. Therefore, no further separation was carried out.


To optimize all parameters of the direct ELISA protocol, Pierce 96-Well Polystyrene Plates, (ThermoFisher Scientific) were coated overnight at 4° C. with different concentrations of intact virus particles (ZIKV or DENV). The intact virus particles were diluted using 100 mM NaHCO3, pH 9.6, and aliquots of 100 μL were dispensed into each well of the plate using a multichannel pipette. This buffer pH assured a strong hydrophobic binding interaction between polystyrene and virus particles.


Intact particles both ZIKV and DENV were provided by Dr. Watkins group (University of Miami, Dep. of Pathology). The samples were controlled and counted by focus forming assay and RT-PCR, the details were reported in a previous work (Magnani et al., 2017). The Intact particles of ZIKV were inactivated using gamma irradiation. Assay biohazardous steps were carried out according to standard safety procedures.


After coating the plates overnight, the intact virus particles were removed by washing five times with the washing buffer (PBST) 10 mM PBS pH 7.4, 0.1% Tween-20, using an automated plate washer (MultiWash+, Molecular Devices, Sunnyvale, Calif.). Then, the plates were blocked with 200 μL of blocking buffers while shaking at 300 rpm at room temperature. The blocking buffers used were: Pierce™ Protein-Free (PBS) Blocking Buffer (PF), Blocker™ BLOTTO in TBS, SuperBlock™ Blocking Buffer, Blocker™ BSA (1×) in PBS. All blocking buffer were from ThermoFisher Scientific.


After 2 h the blocking buffers were removed using the same washing procedure mentioned above. 100 μL-aliquots of several dilutions of peptides biotinylated in 10 mM PBS pH 7.4 were placed in each well and incubated for 2 h while shaking at 300 rpm at room temperature. After the incubation, the unreacted peptides were removed by using the plate washer with the same settings. Then, 100 μL-aliquots of streptavidin-HRP (ThermoFisher Scientific) at a concentration of 20 ng/mL were added into each well and incubated for 30 min at room temperature without shaking. After the incubation, excess streptavidin-HRP was removed and the wells were washed with the plate washer five times using the washing buffer. Finally, 100 μL-aliquots of the Ultra TMB-ELISA Substrate Solution (ThermoFisher Scientific) were added and after 10 min the reaction was stopped by adding 100 μL aliquots of the TMB stop solution (SeraCare). The emission (450 nm) was read using a microplate reader (Clariostar Optima; BMG Labtech, Ortenberg, Germany).


The corresponding blank signals in triplicates were obtained by using all reagents without peptides. The blank signal was then subtracted to the average absorbance values for triplicate wells of each test.


All Data were processed and fitted using the software XLSTAT Version 2016.02.28451.


Results and Discussion


Docking Simulation


The envelope protein, responsible for virus entry, has very similar structure in all flavivirus. ZIKV differs from other known flavivirus by only ˜10 amino acids that surround the Asn154 glycosylation site found in each of the 180 envelope glycoproteins (Sirohi et al., 2016).


As depicted by FIG. 1A the two arms of the clamp peptide were docked in two different envelope protein binding sites. The first active site box, defining the general space of the protein where peptides are expected to bind, was designed around the amino acid Asn 154 in the SIRE and the amino acids Asn 153, Asn 140 and Asn 134 respectively in the 4UTC, 3N40 and 3I50. The Second active site box was built inside the closest cavity from the first active site, around the amino acid His 323 in the SIRE and the amino acids Val 354, His 331 and His 285 respectively in the 4UTC, 3N40 and 3I50.



FIG. 1B showed the schematic representation of the clamp peptide structure made by two peptide arms of five or six amino acids in length linked by a bridge peptide having as sequence GPGCCGPG (SEQ ID NO: 32). The length in angstrom (1.931 nm) between two peptides docked in the two active sites chosen within the envelope protein was also calculated (FIG. 1C) in order to have an idea of the length needed to link the two arm peptides with a bridge peptide. The bridge peptide chosen (GPGCCGPG; SEQ ID NO: 32) had a length after minimization of 1.956 nm (FIG. 1D), approximately the distance between the two arm peptides. Using this bridge peptide size, the two arms of the clamp peptide should have enough flexibility to bind each of the two active site in synergic combination.


In order to avoid any disulfide bond between the two arms of the clamp peptide, the peptides having sulfur-containing amino acids (cysteine and methionine) were discarded during the semi-combinatorial peptide libraries screening.


The docking process was run in 3 steps. In each step a peptide library was generated by using an incremental construction approach. In every subsequent iteration, a focused library of peptides of increasing complexity, was built on previous iteration results. The first peptide library docked was made by the entire 160 k possible tetrapeptide combinations of the 20 natural amino acids.


The docking program used in this work was based on multi-conformer rigid body docking, therefore ten conformers per peptide were generated to ensure a good compromise between calculation time and accuracy of the output data for this type of ligands (Perez et al., 2013).


From the 5% peptides (8 k tetrapeptides) having the best biding score for each of the two ZIKV binding sites, only 1 k tetrapeptides were selected for the next step.


The criterion of the selection was to choose the peptides inside the top 5% peptides binding the ZIKV active site and concurrently outside the top 5% peptides binding the other flaviviruses binding sites. The meaning of the selection was to maximize the recognition properties of the amino acid motif between the ZIKV binding site and the other flaviviruses binding sites (DENV, West Nile and Chikungunya). The 5% was selected as cutoff because in all simulations, this value delimited the zone of the curve in which the steeper slope change was observed (FIG. 2A).


This criterion was applied to the other steps to select penta, hexa and heptapeptides. FIG. 2A depicted the typical distributions of scores obtained in the simulations. The curves obtained had similar gaussian distributions. Score values comprised within the range from 20 to −10 kcal/mol in all simulations. All docking runs had approximately 5% of the complexes with higher scores and 5% with worse scores, both well separated from the rest of the population.


The score values were calculated using chemgauss4 scoring function and, thus, lower values represented higher protein-peptide affinity.


The second step was the generation of the pentapeptide library by inserting each of the 20 natural amino acids in every position of the 1 k tetrapeptides selected in the previous step. As reported in FIG. 2B, the hexapeptide library was built by using the same semi-combinatorial approach carried out in the first step but selecting the best 1 k pentapeptides. A total of 380 k peptides were docked in each active site box.


Table 1 reports the statistical summary of the binding scores calculated for the three libraries of peptides towards the ZIKV envelope protein. The score values were calculated using chemgauss4 scoring function and, thus, lower values represented higher protein-peptide affinity. The peptide size played a critical role for the active site 1 interaction, with lower values decreasing drastically from pentapeptides to hexapeptides, but not for active site 2.












TABLE 1







active site 1
Active site 2



















Tetrapeptides












min
−7.5
−7.0



max
16.5
6.3



Av
0.1
−2.2



median
−0.2
−2.2










Pentapeptides












min
−9.0
−7.0



max
9.6
8.1



Av
−1.0
−0.8



median
−1.1
−0.9










Hexapeptides












min
−5.8
−7.9



max
21.8
26.8



Av
1.9
2.5



median
1.6
2.2







Statistical parameters of the scores behavior (Kcal/mol), obtained using the three peptide libraries docked in the active site 1 (glycosylation binding site) and the active site 2 of the 5IRE ZIKV envelope protein.






These results could be explained considering the steric effects of the peptides within the glycosylation binding pocket. Also, the minimum-maximum dynamic range among the peptide libraries reflected that this behavior becomes relevant for hexapeptides. On the other hand, all peptide libraries showed average and median very close to each other, demonstrating a good symmetry in normal distribution.


It should be noted that the purpose of this work was to use the virtual screening step to select peptides for the specific detection of ZIKV virus among different flaviviruses.


The docking results were used to select the arms of the clamp peptides for the experimental part. This selection was based on the peptide primary structure structural analysis results along with the position in the top ranked peptides and peptide length.


The primary structural analysis of the docking results was carried out to study the occurrence of the amino acids in the top 0.1% ranked peptides that maximized the recognition properties between the ZIKV active site and the other flaviviruses binding sites (DENV, West Nile and Chikungunya).


One peptide with high and one with low occurrence amino acids in primary structure were then chosen within the 0.1% top ranked peptides binding each of the two envelope protein active sites. The four peptides were selected from both penta and hexapeptide libraries, resulting in a total of 8 peptides, four pentapeptides and four hexapeptides.


Table 2 reports the results of the amino acid occurrence (%) in the primary structure of the eight peptides selected to build the clamp peptides. The occurrence was calculated counting the recurrence of each amino acid in the relative position (five positions for pentapeptides and six positions for hexapeptides) in the top 0.1% ranked peptides binding the active site 1 and 2 of the ZIKV envelope protein. The best occurrence amino acids were also reported, but peptides having all best occurrence amino acids were not present or were in the bottom of the 0.1% top rank peptides.









TABLE 2





Amino acid occurrence in the top 160 ranked peptides (%)
























1P
2P
3P
4P
5P
Average





Active
SWPGQ (24)
24.4
13.8
22.5
34.4
1.3
19.3%


site 1
LRGHA (25)
11.3
8.1
21.9
21.3
7.5
14.0%




best


24.4


14.4


31.3


34.4


18.8


24.6%





occurrence





AA: SMAGG




(37)


Active
WPHTQ (16)
58.8
63.1
15.0
4.4
10.0
30.3%


site 2
AGRRP (20)
5.0
4.4
9.4
6.3
28.1
10.6%




best


58.8


63.1


20.0


13.1


28.1


36.6%





occurrence





AA: WPFFP




(38)




















1P
2P
3P
4P
5P
6P
Average





Active
KRNATP (26)
10.0
6.3
28.8
56.9
34.4
41.9
29.7%


site 1
KTDAYS (27)
10.0
10.0
3.1
56.9
2.5
3.8
14.4%




best


14.4


11.9


28.8


56.9


34.4


41.9


31.4%





occurrence





AA: GPNATP




(39)


Active
WPWIGT (18)
75.0
80.0
32.5
13.1
35.6
8.8
40.8%


site 2
MDSPIK (22)
1.3
0.6
1.3
2.5
1.9
2.5
1.7%




best


75.0


80.0


32.5


27.5


35.6


18.8


44.9%





occurrence





AA: WPWFGP




(40)





Analysis of the amino acid occurrence (%) in the primary structure of the eight peptides selected to build the eight clamp peptides. The occurrence was calculated counting the recurrence of each amino acid in the relative position (five positions for pentapeptides and six positions for hexapeptides) in the top 0.1% ranked peptides binding the active site 1 and 2 of the ZIKV envelope protein. The best occurrence amino acids were also reported (in italic) along with the average (Av) percentage of the occurrence for each peptide.


SEQ ID NO: noted in ( ).






In the active site 1 of ZIKV envelop protein (glycosylation active site), the pentapeptide SWPGQ (SEQ ID NO: 24) and hexapeptide KRNATP (SEQ ID NO: 26) had almost all amino acids with high occurrence with some exception, the glutamine in fifth position for SWPGQ (SEQ ID NO: 24) and lysine in second position for KRNATP (SEQ ID NO: 26). The average in percentage of the occurrence (19.3% and 29.7%) was the highest of the top 0.1% ranked peptides in their respective libraries.


The other pentapeptide LRGHA (SEQ ID NO: 25) had amino acids with about half percentage of occurrence when compared to the top one in almost all the five positions. On the other hand, the hexapeptide KTDAYS (SEQ ID NO: 27) showed and alternate low and high occurrence percentage.


The penta and hexapeptides selected from the active site 2 docking, had similar high and low average occurrence amino acids in primary structure. Remarkably, the hexapeptides WPWIGT (SEQ ID NO: 18) and MDSPIK (SEQ ID NO: 22) had respectively the highest and lowest amino acids occurrence (40.8% and 1.7%) of the peptides selected to build the clamp peptides.


Table 3 shows the relative docking score position in the corresponding libraries of the eight peptides chosen to build the arms of the clamp peptides tested in the experimental evaluation. The ranking scores between the ZIKV sites and the other flaviviruses binding sites were for all peptides different enough to expect a ZIKV selective binding. Nevertheless, only three peptides ranked in the first 10 best peptides in binding ZIKV, highlighting strong similarities between the flaviviruses glycosylation sites. It should be noted that in all peptide libraries the simulated binding energy decreased exponentially in the top 1% best ranked peptides, in fact, a decrease of at least 20% in the binding score is observed for the 100th peptide position.















TABLE 3







Peptide


West




(SEQ ID NO:)
ZIKV
DENV
Nile
Chikungunya



















Active site 1




Docking score rank













LRGHA (25)
53
74900
69645
71784



SWPGQ (24)
4
55575
27840
47111



KRNATP (26)
16
85123
100716
74427



KTDAYS (27)
125
95558
110693
86882










Active site 2




Docking score rank













WPHTQ (16
9
47135
43238
45072



AGRRP (20)
13
29265
51031
18894



WPWIGT (18)
1
21334
9821
18311



MDSPIK (22)
46
10080
40000
7831







Relative docking score position of the four pentapeptides and four hexapeptides selected for building the eight clamp peptides. The ranking obtained from the two active sites of the ZIKV envelope protein was compared to the one obtained using the other three flaviviruses envelope proteins.






The four pentapeptides and four hexapeptides were then combined to build eight clamp peptides. At this stage a mix between penta and hexapeptides was avoided in order to understand the contribution of the peptide length in the experimental responses.


Table 4 reports the physicochemical properties of the peptides selected for experimental evaluation. The peptides were the eight clamp peptides built using the combination of the penta or hexapeptides, and the mono-arm peta and hexapeptides binding the ZIKV envelope protein glycosylation binding site. Cysteine was inserted in the bridge of clamp peptides and at the N-terminus of mono-arm peptides to bind the maleimide-PEG2-biotin used to label each of the peptides with the signal amplifier streptavidin-HRP.
















TABLE 4 





Active

Active

Iso-
net




site
Bridge
site

Point
charge
Water



2*
Peptide*
1*
Label
pH
pH7
Sol
MW























WPHTQ

GPGCCG

PGSWPGQ

C1
6.9
0.0
poor
1852



(16)

(32)

(24)












WPHTQ

GPGCCG

PGLRGHA

C2
8.1
1.1
poor
1831



(16)

(32)

(25)












WPWIGT

GPGCCGPG

KRNATP

C3
8.9
1.9
poor
2055



(18)

(32)

(26)












WPWIGT

GPGCCGPG

KTDAYS

C4
5.8
−0.1
poor
2053



(18)

(32)

(27)












AGRRP

GPGCCGPG

SWPGQ

C5
12.1
2.0
good
1739



(20)

(32)

(24)












AGRRP

GPGCCGPG

LRGHA

C6
10.6
3.0
good
1718



(20)

(32)

(25)












MDSPIK

GPGCCGPG

KRNATP

C7
8.8
1.9
good
1986



(22)

(32)

(26)












MDSPIK

GPGCCGPG

KTDAYS

C8
5.9
−0.1
good
1984



(22)

(32)

(27)














C-SWPGQ

P1
3.0
−0.1
poor
676





(33)














C-LRGHA

P2
9.2
1.0
good
655





(34)














C-KRNATP

X1
10.5
1.9
good
788





(35)














C-KTDAYS

X2
5.9
−0.1
good
786





(36)






Physicochemical properties of the eight peptides selected for experimental part.


A cysteine was added to the N terminus of each mono-arm peptide to link the maleimide-PEG2-biotin.


*SEQ ID NOs: are noted in ( ).






The experimental analysis was performed in PBS at pH 7.4. Therefore, the physicochemical properties were focused on water solubility and net charge at pH 7. The first four clamp peptides and one of the mono-arm pentapeptide had poor water solubility due to the ratio of the hydrophobic amino acids, but when they were used at micromolar concentration were able to be dissolved in PBS. Five clamp peptides and two mono-arm peptides had a significant amount of positively charged amino acids resulting in a positive net charge at pH 7. Due to the presence of the polar amino acids the other peptides had a slightly negative net charge at pH 7.


Moreover, to highlight the positive or negative charges inside the peptide, the pH of the isoelectric point of each peptide was also reported. Interestingly, the majority of peptides selected had positively charged amino acids improving the possibility to interact with negative charges in the three-dimensional structure of both ZIKV binding sites selected.


Experimental Results


The eight clamp peptides and the four mono-arm peptides selected, were tested vs intact ZIKV particles by using a direct ELISA. The peptides were biotinylated by using the maleimide-PEG2-biotin reagent that reacts with the sulfhydryl group of the cysteine efficiently and specifically by forming a stable thioether bond. The antibody 4G2 hybridoma mouse IgG2a was used as the reference and employed in combination with an anti-mouse IgG conjugated to HRP.


All analytical parameters involved in the development of ELISA were optimized by using 96-well plates coated with triplicate 10-fold serial dilutions of intact ZIKV particles. The results were reported in Table 5.























TABLE 5



















AB



C1
C2
C3
C4
C5
C6
C7
C8
P1
P2
X1
X2
4G2






























Blocking

BT
BT
BT
BT
PF
PF
PF
PF
PF
PF
PF
PF
PF


Incubation

PBST
PBST
PBST
PBST
PBST
PBST
PBST
PBST
PBS
PBS
PBS
PBS
PBST


buffer


FPLR
(log[ZIKV],
5-7
5-7
4-7
6-7
6-7
4-7
4-7
5-7
7-8
5-7
5-7
6-8
6-8


Dynamic
copies/mL)


Range


LOD
(log[ZIKV],
4.8
4.5
3.5
5.7
5.8
3.5
3.7
4.7
6.8
4.5
4.8
5.8
5.8



copies/mL)


FPLR C50
(log[ZIKV],
6.3
6.2
5.3
6.3
6.4
5.2
5.8
6.2
nd
6.2
6.1
6.1
6.2



copies/mL)


FPLR slope
ΔA/
33.2
25.4
8.7
32.8
25.6
10.1
5.3
33.9
nd
19.9
20.8
27.7
22.3



(log[ZIKV])


FPLR
ΔA
0.43
0.55
0.67
0.40
0.17
0.53
0.95
0.82
nd
0.94
0.98
0.91
0.81


maximum


FPLR
ΔA
0.03
0.05
0.05
0.02
0.01
0.05
0.01
0.03
nd
0.04
0.03
0.01
0.04


minimum


FPLR R{circumflex over ( )}2

0.98
1.00
0.99
1.00
0.97
0.99
0.99
0.99
nd
0.99
1.00
1.00
0.99


Peptide
(μM)
2
2
0.5
2
5
0.5
5
5
20
20
20
20
1


Concen-













□g/ml


tration


Intra-day
CV(%)
<5
<5
<7
<5
<4
<7
<4
<4
<5
<5
<5
<5
<10


repro-


ducibility


Inter-day and
CV(%)
<12
<12
<15
<12
<10
<15
<10
<10
<10
<10
<10
<10
nd


batch-to-


batch repro-


ducibility


Long-term
(Month)
>1
>1
>1
>1
>1
>1
>1
>1
>1
>1
>1
>1
nd


stability


Assay time
(h)
5
5
5
5
5
5
5
5
5
5
5
5
8


after Plate


Coating





Optimized experimental parameters of the direct ELISA assay for the eight clamp peptides, the four mono-arm peptides and a commercial antibody (4G2).


BT = BLOTTO blocking buffer;


PF = protein free blocking buffer;


PBS = 10 mM phosphate buffer saline pH 7.4;


PBST= 10 mM PBS pH 7.4, 0.1% Tween-20.


FLRP = Four Parameter Logistic Regression






To minimize nonspecific binding, four blocking agents (PF, BLOTTO, SuperBlock™ Blocking Buffer, Blocker™ BSA) were tested. All blocking agents had very low background signal. For hydrophilic peptides the lowest background signal was achieved using PF that gave the best performances also using the antibody 4G2. For hydrophobic peptides, the blocker BLOTTO showed better performance, except for mono-arm peptide P1.


For clamp peptides and antibody, the surfactant agent tween 20 at 0.1% was necessary in the incubation step. No longer than one hour was necessary for peptide incubation, a longer time increased both the overall signal generated by the binding event and the background signal. Shacking during incubation improved the signal to noise ratio.


The optimal concentration of peptide was determined by coating clear 96-well plates with a solution of 10{circumflex over ( )}7 copies/mL of intact ZIKV particles. Concentrations of peptide, from 0.1 to 50 μM, diluted in 10 mM PBS pH 7.4 were added to wells of the microplates coated with intact ZIKV particles. For mono-arm peptides, larger concentrations than 20 □M did not increase the assay sensitivity.


Clamp peptides showed higher sensitivity than mono-arm peptides. Clamp peptides C5, C7 and C8 showed the best performances when used at 5 μM. The clamp C1, C2 and C4 at concentration of 2 μM did the best signal to noise ratio. Impressively, clamp peptides C3 and C6 had be used at concentration of 0.5 μM to have the best results, highlighting the remarkable high sensitivity of those peptides in this kind of assay.


Thus, the peptide concentrations reported in Table 5 were used to estimate the dynamic range and the LOD of the assay by using 10-fold serial dilutions of intact ZIKV particles from 10{circumflex over ( )}1 to 10{circumflex over ( )}8 copies/mL.


The results had a sigmoidal ZIKV particles concentration response and the calibration curves were obtained by plotting the delta absorbance (after blank signal subtraction) against the log of ZIKV particles concentration and fitting the experimental data with a four-parameter logistic function (FPLR).


The regression parameters of the assay were reported in Table 5 and the sigmoidal trend in FIG. 3. The LOD was interpolated from the calibration curves using LOD=SB+3×SDB where SB and SDB were the average and the standard deviation of the blank measurements, respectively.


Dose-response curves generated with all peptides and the antibody had at least two-order of magnitude dynamic range except for peptide P1, which had just a one order of magnitude dynamic range.


The peptide based assay using C3, C6, and C7 showed three-order of magnitude dynamic range and lower detection limits with dynamic range starting from 10{circumflex over ( )}4 copies/mL one or two order magnitude lower than the others peptides or antibody based assay. The better performance in binding ZIKV intact particles by those three clamp peptides was also highlighted by the FPLR C50 parameter. The dose-response performance of the assay was reproducible over a month (RSD lower than 15%), demonstrating that the peptides had high stability and reproducibility.


The cross-reactivity among Flaviviruses is a key parameter to be tested for this assay. Using the same ELISA protocol, the three clamp peptides (C3, C6 and C7) with higher sensitivity versus the intact ZIKV particles where employed to test the ability to discriminate ZIKV from the three serotypes of DENV (DENV-1, -2 and -3). The cross reactivity performances were compared with that obtained using the mono-arm peptides P2 and X1, that showed the best analytical parameters within the mono-arm peptides.


The results shown in FIG. 4 were obtained by coating clear 96-well plates with a solution of 10{circumflex over ( )}5 copies/mL of intact virus particles. At this concentration, all three peptides showed slight cross-reactivity against the DENV. Clamp peptides C3 and C7 had the higher DENV/ZIKV signal ratio with around 70% signal decrease for all DENV serotypes. Clamp Peptide C6 showed slight cross reactivity with DENV-2 and DENV-3 with only 45% and 55% of signal decrease respectively. The three clamp peptides clearly discriminated between the two flavivirus species. At this concentration, mono-arm peptide assays had the ZIKV delta absorbance signals statistically comparable to the signals obtained using DENV.


Nevertheless, it should be highlight that increasing the coating concentration of the virus to 10{circumflex over ( )}6 copies/mL the ZIKV and DENV analytical signals were statistically equivalent, losing, for the clamp peptides, the discrimination between ZIKV and DENV.


Usually, the presence of ZIKV in affected bodies is detected in biological fluids. Therefore, the analytical sensitivity of the selected peptides was tested in two biological matrices, namely, urine and serum. The matrix effect was investigated to understand how real biological fluids could modify the binding efficiency of the peptides.



FIG. 4 depicts the ELISA data using solutions of peptides with or without the urine and serum obtained coating clear 96-well plates with 10{circumflex over ( )}5 and 10{circumflex over ( )}6 copies/mL of intact ZIKV particles. Urine and serum were 1:1 and 1:10 diluted, respectively, with a concentrated peptide PBS solution (10 mM PBS, pH 7.4) to obtain a peptide final concentration of 0.5 μM for C3 and C6, 5 μM for C7 and 20 μM for mono-arm peptides (P2 and X1).


Clamp peptide C3 showed a better performance in urine than serum, particularly for 10{circumflex over ( )}5 copies/mL. Clamp peptide C6 had a strong decrease in the signal generated at both 10{circumflex over ( )}5 or 10{circumflex over ( )}6 copies/mL in both urine and serum. Clamp peptide C7 exhibited the best performance among the three clamp peptides, having higher signals in serum than in urine. All clamp peptides lost at the least one order of magnitude signal detecting ZIKV in urine or serum, except for C7 in serum, having a distinct analytical signal also at 10{circumflex over ( )}5 copies/mL of ZIKV.


Mono-arm peptides lost completely the signal at 10{circumflex over ( )}5 copies/mL of ZIKV, starting to detect ZIKV particles in both urine or serum, from 10{circumflex over ( )}6 copies/mL using P1 and from 10{circumflex over ( )}7 copies/mL using X1 (data not showed).


It should be highlighted that when a body is under a ZIKV infection the level of flavivirus concentration in urine can be up to 10{circumflex over ( )}6.9 copies/mL (Campos et al., 2016; Pawley et al., 2019).


CONCLUSIONS

The semi combinatorial virtual strategy to design clamp peptides using the two flavivirus active sites as a binding target has shown to have the potential for designing antibody mimetics for the selective detection of ZIKV.


The ELISA assay platform developed for testing the newly designed clamp peptides offered the possibility to optimize in short time the experimental conditions for evaluation of the eight clamp peptides and four mono-arm peptides chosen from the most promising ones yield by the in-silico studies. The three clamp peptides with better performances to detect ZIKV had shown to have also semi-selective properties when tested against DENV. The matrix-effect was also investigated, by testing the response of the peptides in physiological matrices, i.e., urine and serum. We observed that the matrix affected the assay performance by decreasing the detection limits by one order of magnitude, albeit still having a distinct analytical signal starting from 10{circumflex over ( )}6 copies/mL, the concentration of ZIKV in acute infection.


This work represents a new methodology for the selection of tailor-made clamp peptides, rationalizing the way to choose receptors with high binding ability among thousands of potential compounds that can be employed in biotechnology, medical, and a variety of analytical applications. Taking advantage of the fast progress in computing, we envision that it will be possible to simulate in short time the clamp peptides having even more complex shapes with better selectivity and less cross-reactivity.


REFERENCES



  • Acebes et al., ACS Catalysis 6(3), 1624-1629 (2016).

  • Barba-Spaeth et al., Nature 536(7614), 48. (2016).

  • Bunker et al., Biochimica et Biophysica Acta (BBA)-Biomembranes 1858(10), 2334-2352 (2016).

  • Campos et al., Journal of Clinical Virology 77, 69-70 (2016).

  • Do Thi Hoang Kim et al., Theranostics 8(13), 3629 (2018).

  • Gong et al., Yi Chuan 32(6), 548-554 (2010).

  • Hawkins et al., Journal of Chemical Information and Modeling 52(11), 2919-2936. doi: 10.1021/ci300314k.

  • Hawkins et al., Journal of Chemical Information and Modeling 50(4), 572-584. doi: 10.1021/ci100031x. (2010)

  • Heffron et al., PLoS neglected tropical diseases 12(11), e0006903 (2018).

  • Hwang et al., Biosensors and Bioelectronics 87, 164-170 (2017).

  • Ioos et al., Medecine et maladies infectieuses 44(7), 302-307 (2014).

  • Kelley et al., POSIT: flexible shape-guided docking for pose prediction. Journal of chemical information and modeling 55(8), 1771-1780 (2015).

  • Li et al., Electrochemical aptamer-based sensors for food and water analysis: A review. Analytica chimica acta. (2018)

  • Lin et al., Journal of Chromatography A 1446, 34-40 (2016).

  • Macalino et al., Archives of pharmacal research 38(9), 1686-1701 (2015).

  • Magnani et al., Science translational medicine 9(410), eaan8184 (2017).

  • Mascini et al., Biosensors and Bioelectronics 123, 124-130 (2019).

  • Mascini et al., Talanta 167, 126-133 (2017).

  • Michaeli et al., The Journal of Immunology 201(11), 3383-3391 (2018).

  • OEDocking. version 3.0.0. OpenEye Scientific Software, Santa Fe, N. Mex. http://www.eyesopen.com.

  • OMEGA. version 2.4.6. OpenEye Scientific Software, Santa Fe, N. Mex. http://www.eyesopen.com.

  • Palzkill et al., U.S. patent application Ser. No. 14/777,714), (2018)

  • Pawley et al., ACS Omega 4(4), 6808-6818 (2019).

  • Perez et al., Peptides trapping dioxins: a docking-based inverse screening approach. Journal of Chemistry 2013.

  • Pichon et al., Anal Bioanal Chem 407(3), 681-698. doi: 10.1007/s00216-014-8129-5 (2015).

  • Priyamvada et al., Proceedings of the National Academy of Sciences 113(28), 7852-7857 (2016).

  • Singh et al., Journal of medicinal chemistry 59(8), 3920-3934 (2016).

  • Sirohi et al., Science 352(6284), 467-470 (2016).

  • Stettler et al., Science 353(6301), 823-826 (2016).

  • Stobiecka et al., Chemical Papers 69(1), 62-76 (2015).

  • SZYBKI. version 1.5.7. OpenEye Scientific Software, Santa Fe, N. Mex. http://www.eyesopen.com.

  • Tambunan et al., Jurnal Teknologi 78(4-3) (2016).

  • Tang et al., Talanta 146, 55-61 (2016).

  • VIDA. version 4.1.1. OpenEye Scientific Software, Santa Fe, N. Mex. http://www.eyesopen.com.

  • Weaver et al., Antiviral research 130, 69-80 (2016).

  • Xu et al., Nature medicine 24(6), 857 (2018)

  • Yu et al., Annual Review of Analytical Chemistry 10, 293-320 (2017).

  • Yuan et al., Wiley Interdisciplinary Reviews: Computational Molecular Science 7(2), e1298 (2017).

  • Zhao et al., Structural basis of Zika virus-specific antibody protection. Cell 166(4), 1016-1027 (2016).



All references, including publications, patent applications, and patents, cited herein are hereby incorporated by reference to the same extent as if each reference were individually and specifically indicated to be incorporated by reference and were set forth in its entirety herein.


The use of the terms “a” and “an” and “the” and similar referents in the context of describing the disclosure (especially in the context of the following claims) are to be construed to cover both the singular and the plural, unless otherwise indicated herein or clearly contradicted by context. The terms “comprising,” “having,” “including,” and “containing” are to be construed as open-ended terms (i.e., meaning “including, but not limited to,”) unless otherwise noted.


Recitation of ranges of values herein are merely intended to serve as a shorthand method of referring individually to each separate value falling within the range and each endpoint, unless otherwise indicated herein, and each separate value and endpoint is incorporated into the specification as if it were individually recited herein.


All methods described herein can be performed in any suitable order unless otherwise indicated herein or otherwise clearly contradicted by context. The use of any and all examples, or exemplary language (e.g., “such as”) provided herein, is intended merely to better illuminate the disclosure and does not pose a limitation on the scope of the disclosure unless otherwise claimed. No language in the specification should be construed as indicating any non-claimed element as essential to the practice of the disclosure.


Preferred embodiments of this disclosure are described herein, including the best mode known to the inventors for carrying out the disclosure. Variations of those preferred embodiments may become apparent to those of ordinary skill in the art upon reading the foregoing description. The inventors expect skilled artisans to employ such variations as appropriate, and the inventors intend for the disclosure to be practiced otherwise than as specifically described herein. Accordingly, this disclosure includes all modifications and equivalents of the subject matter recited in the claims appended hereto as permitted by applicable law. Moreover, any combination of the above-described elements in all possible variations thereof is encompassed by the disclosure unless otherwise indicated herein or otherwise clearly contradicted by context.

Claims
  • 1. A method implemented by a processor in a computer for iteratively designing a linear clamp peptide that binds to two different binding sites of a target protein, wherein the linear clamp peptide comprises the structure A1-B-A2 wherein A1 is a first peptide arm of the linear clamp peptide binds to a first binding site of a target protein, A2 is a second peptide arm of the linear clamp peptide binds to a second binding site of the target protein, and B is a bridge peptide which links A1 to A2, the iterative designing method comprising the steps of: (a) determining a binding score of each tetrapeptide of a first peptide library for the first binding site of the target protein, wherein the first peptide library comprises a full combinatorial set of 160 K tetrapeptides having a combination of four amino acids of the twenty naturally-occurring amino acids,(b) identifying the sequences of the tetrapeptides having a binding score for the first binding site of the target protein which meets a first threshold, wherein the first threshold is a score of binding between each tetrapeptide and a first active site of the target protein;(c) determining a binding score of each pentapeptide of a second peptide library for the first binding site of the target protein, wherein the second peptide library comprises a set of pentapeptides having a combination of five amino acids comprising the amino acids of the sequence of a tetrapeptide identified in step (b) and one of the twenty naturally-occurring amino acids added as the first amino acid or the fifth amino acid of the pentapeptide or between two amino acids of the sequence of the tetrapeptide,(d) identifying the sequences of the pentapeptides having a binding score for the first binding site of the target protein which meets a second threshold, wherein the second threshold is a score of binding between each pentapeptide and a first active site of the target protein,(e) determining a binding score of each hexapeptide of a third peptide library for the first binding site of the target protein, wherein the third peptide library comprises a set of hexapeptides having a combination of six amino acids comprising the amino acids of the sequence of a pentapeptide identified in step (d) and one of the twenty naturally-occurring amino acids added as the first amino acid or the sixth amino acid of the hexapeptide or between two amino acids of the sequence of the pentapeptide,(f) identifying the sequences of the hexapeptides having a binding score for the first binding site of the target protein which meets a third threshold, wherein the third threshold is a score of binding between each hexapeptide and a first active site of the target protein,(g) repeating steps (a). (c) and (e) to determine a binding score for the second binding site of the target protein for each tetrapeptide of the first peptide library, for each pentapeptide of the second peptide library, and for each hexapeptide of the third peptide library;(h) repeating steps (b), (d), and (f) to identify the sequences of the tetrapeptides, pentapeptides and hexapeptides having a binding score for the second binding site of the target protein which meets a first, second, and third threshold, wherein the first threshold is a score of binding between each tetrapeptide and a second active site of the target protein, wherein the second threshold is a score of binding between each pentapeptide and a second active site of the target protein, and wherein the third threshold is a score of binding between each hexapeptide and a second active site of the target protein;(i) selecting the A1 peptide arm based on peptides identified as having a binding score for the first binding site of the target protein that meets the first, second and third threshold,(j) selecting the A2 peptide arm based on peptides identified as having a binding score for the second binding site of the target protein that meets the first, second and third threshold,(k) generating the linear clamp peptide, wherein the linear clamp peptide comprises the A1 peptide selected in step (i) and the A2 peptide selected in step (i).
  • 2. The method of claim 1, comprising generating the first peptide library, the second peptide library, the third peptide library, and/or the fourth peptide library.
  • 3. The method of claim 1, wherein the first peptide library comprises 160,000 tetrapeptides.
  • 4. The method of claim 1, wherein the first threshold is the top 5% of binding scores, and wherein 8000 sequences of tetrapeptides are identified upon step (b).
  • 5. The method of claim 1, wherein the first threshold is the top 5% of binding scores and a binding score outside the top 5% of binding scores for a different target protein, and wherein 1000 sequences of tetrapeptides are identified upon step (b).
  • 6. The method of claim 1, wherein the second threshold is the top 5% of binding scores, and wherein the second threshold further comprises a binding score outside the top 5% of binding scores for a different target protein.
  • 7. The method of claim 1, wherein the third threshold is the top 5% of binding scores, and wherein the third threshold further comprises a binding score outside the top 5% of binding scores for a different target protein.
  • 8. The method of claim 1, wherein binding scores are calculated using a docking scoring function.
  • 9. The method of claim 1, wherein the binding scores for more than 350,000 peptides are determined for each of the first binding site and the second binding site.
  • 10. The method of claim 1, further comprising determining the length of B of the clamp peptide by measuring the distance between a peptide bound to the first binding site of the target protein and a peptide bound to the second binding site of the target protein.
  • 11. The method of claim 1, wherein binding scores are determined using a molecular docking program.
  • 12. The method of claim 11, wherein the molecular docking program is based on multi-conformer rigid body docking, which evaluates 10 conformers per peptide.
  • 13. The method of claim 8, wherein the docking scoring function is chemgauss4.
  • 14. A method of manufacturing a linear clamp peptide that binds to two different binding sites of a target protein, wherein the linear clamp peptide comprises the structure A1-B-A2 wherein A1 is a first peptide arm that binds to a first binding site of a target protein, A2 is a second peptide arm that binds to a second binding site of the target protein, and B is a bridge peptide which links A1 to A2, said method comprising iteratively designing the first peptide arm and the second peptide arm of the clamp peptide according to the method of claim 1 and joining the first peptide arm to the second peptide arm with a bridge peptide, B.
CROSS-REFERENCE TO RELATED APPLICATION(S)

This application claims priority to U.S. Provisional Application No. 62/684,960, filed on Jun. 14, 2018, the contents of which are incorporated herein by reference.

US Referenced Citations (38)
Number Name Date Kind
4946778 Ladner et al. Aug 1990 A
5225539 Winter Jul 1993 A
5403484 Ladner et al. Apr 1995 A
5545806 Onberg et al. Aug 1996 A
5565325 Blake Oct 1996 A
5569825 Lonberg et al. Oct 1996 A
5571698 Ladner et al. Nov 1996 A
5585089 Queen et al. Dec 1996 A
5639641 Pedersen et al. Jun 1997 A
5693761 Queen et al. Dec 1997 A
5693762 Queen et al. Dec 1997 A
5702892 Mulligan-Kehoe Dec 1997 A
5714352 Jakobobits et al. Feb 1998 A
5780279 Matthews et al. Jul 1998 A
5821047 Garrard et al. Oct 1998 A
5824520 Mulligan-Kehoe Oct 1998 A
5837500 Ladner et al. Nov 1998 A
5855885 Smith et al. Jan 1999 A
5858657 Winter et al. Jan 1999 A
5871907 Winter et al. Feb 1999 A
5969108 McCafferty et al. Oct 1999 A
6057098 Buechler et al. May 2000 A
6225447 Winter et al. May 2001 B1
6265150 Terstappen et al. Jul 2001 B1
6551592 Lindhofer et al. Apr 2003 B2
6699843 Pietras et al. Mar 2004 B2
7355008 Stavenhagen et al. Apr 2008 B2
7381408 Mezo et al. Jun 2008 B2
9885050 Koide Feb 2018 B2
20020197266 Debinski Dec 2002 A1
20050282233 Eriksson et al. Dec 2005 A1
20090081230 Lanzavecchia et al. Mar 2009 A1
20110171241 Dix et al. Jul 2011 A1
20120020957 Lanzavecchia Jan 2012 A1
20150355192 Sun Dec 2015 A1
20170336404 Ali Nov 2017 A1
20180179519 Wang et al. Jun 2018 A1
20200135299 Wang Apr 2020 A1
Foreign Referenced Citations (8)
Number Date Country
105954512 Sep 2016 CN
0239400 Aug 1994 EP
2188638 Oct 1987 GB
2005087812 Sep 2005 WO
2010051007 May 2010 WO
2016022071 Feb 2016 WO
2016069245 May 2016 WO
2017197477 Nov 2017 WO
Non-Patent Literature Citations (103)
Entry
Winter et al., Man-made antibodies, Nature, 349:293-299 (1991).
Woodlock et al., Active specific immunotherapy for metastatic colorectal carcinoma: phase I study of an allogeneic cell vaccine plus low-dose interleukin-1 alpha, J. Immunother., 22:251-259 (1999).
Xu et al., Epitope-based vaccine design yields fusion peptide-directed antibodies that neutralize diverse strains of HIV-1, Nature Medicine, 24(6):857-867 (2018).
Yu et al., Beyond Antibodies as Binding Partners: The Role of Antibody Mimetics in Bioanalysis, Annual Review of Analytical Chemistry, 10:293-320 (2017).
Yuan et al., Using PyMOL as a platform for computational drug design, Computational Molecular Science, 7:e1298 (2017).
Zhao et al., Structural Basis of Zika Virus-Specific Antibody Protection, Cell, 166:1016-1027 (2016).
Krammer et al., LigScore: a novel scoring function for predicting binding affinities, J. Molecular Graphics and Modelling, 23(5):395-407 (2005).
Li et al., Electrochemical aptamer-based sensors for food and water analysis: A review, Analytica. Chimica. Acta., (2018).
Lin et al., Selective dispersive solid phase extraction-chromatography tandem mass spectrometry based on aptamer-functionalized UiO-66-NH2 for determination of polychlorinated biphenyls, Journal of Chromatography A, 1446:34-40 (2016).
Logean et al., Customized versus universal scoring functions: application to class I MHC-peptide binding free energy predictions, Bioorganic and Medicinal Chem Lett., 11(5):675-679 (2001).
Loos et al., Current Zika virus epidemiology and recent epidemics, Medicine ET Maladies Infectieuses, 44(7):302-307 (2014).
Macalino et al., Role of computer-aided drug design in modern drug discovery, Archives of Pharmacal. Research, 38(9):1686-1701 (2015).
Magnani et al., A human inferred germline antibody binds to an immunodominant epitope and neutralizes Zika virus, PLoS Negl. Trop. Dis., 11:39-42:1-17, e0005655 (2017).
Magnani et al., Neutralizing human monoclonal antibodies prevent Zika virus infection in macaques, Science Translational Medicine, 9(410):eaan8184 (2017).
Marvin et al., Recombinant approaches to IgG-like bispecific antibodies, Acta. Pharmacologica. Sinica., 26:649-658 (2005).
Mascini et al., Hairpin DNA-AuNPs as molecular binding elements for the detection of volatile organic compounds, Biosensors and Bioelectronics, 123:124-130 (2019).
Mascini et al., Selective solid phase extraction of JWH synthetic cannabinoids by using computationally designed peptides, Talanta, 167:126-133 (2017).
McEnaney et al., Chemically Synthesized Molecules with the Targeting and Effector Functions of Antibodies, J. Am. Chem. Soc., 136(52):18034-18043 (2014).
Michaeli et al., Computationally Designed Bispecific MD2/CD14 Binding Peptides Show TLR4 Agonist Activity, The Journal of Immunology, 201(11):3383-3391 (2018).
Morrison et al., Chimeric human antibody molecules: mouse antigen-binding domains with human constant region domains, Proc. Natl. Acad. Sci., 81:6851-6855 (1984).
Neuberger et al., Recombinant antibodies possessing novel effector functions, Nature, 312:604-608 (1984).
O'Hara et al., Ligand binding assays in the 21st century laboratory: recommendations for characterization and supply of critical reagents, AAPS J., 14(2):316-328 (2012).
OE Docking. version 3.0.0. Open Eye Scientific Software, Santa Fe, NM. http://www.eyesopen.com.
Omega. version 2.4.6. OpenEye Scientific Software, Santa Fe, NM. http://www.eyesopen.com.
Orlandi et al., Cloning immunoglobulin variable domains for expression by the polymerase chain reaction, Proc. Natl. Acad. Sci., 86:3833-3837 (1989).
Pawley et al., Highly Sensitive and Selective Direct Detection of Zika Virus Particles in Human Bodily Fluids for Accurate Early Diagnosis of Infection, ACS Omega, 4(4):6808-6818 (2019).
Pedersen et al., Comparison of Surface Accessible Residues in Human and Murine Immunoglobulin Fv Domains: Implication for Humanization of Murine Antibodies, Journal of Molecular Biology, 235:959-973 (1994).
Perez et al., Peptides trapping dioxins: a docking-based inverse screening approach, Journal of Chemistry, 2013.
Pichon et al., Aptamer-based-sorbents for sample treatment—a review, Anal. Bioanal. Chem., 407(3):681-698 (2015).
Priyamvada et al., Human antibody responses after dengue virus infection are highly cross-reactive to Zika virus, Proceedings of the National Academy of Sciences, 113(28):7852-7857 (2016).
Rajkovic et al., Immunoquantitative real-time PCR for detection and quantification of Staphylococcus aureus enterotoxin Bin foods, Applied and Environmental Microbiology, 72(10):6593-6599 (2006).
Raveh et al., Rosetta FlexPepDock ab-initio: simultaneous folding, docking and refinement of peptides onto their receptors, PLoS One, 6(4):e18934 (2011).
Reiter et al., Engineering interchain disulfide bonds into conserved framework regions of Fv fragments: improved biochemical characteristics of recombinant immunotoxins containing disulfide-stabilized Fv, Protein Engineering, 7:697-704 (1994).
Roder et al., The EBV-hybridoma technique, Methods in Enzymology, 121:140-167 (1986).
Roque et al., Antibodies and genetically engineered related molecules: production and purification, Biotechnology Progressg., 20(3):639-54 (2004).
Routledge et al., The effect of aglycosylation on the immunogenicity of a humanized therapeutic CD3 monoclonal antibody, Transplantation, 60:847-853, (1995).
Sankaranarayanan et al., Broadly Neutralizing Antibodies for therapy of viral Infections, Antibody Tech. Journal, 6:1-15 (2016).
Shields et al., High resolution mapping of the binding site on human IgG1 for Fc gamma RI, Fc gamma RII, Fc gamma RIII, and FcRn and design of IgG1 variants with improved binding to the Fc gamma R, J. Biol. Chem., 276:6591-6604 (2001).
Shukla et al., Rapid Detection Strategies for the Global Threat of Zika Virus: Current State, New Hypotheses, and Limitations, Frontiers in Microbiology, Oct. 24, 2014 (Oct. 24, 2016), vol. 7, Art. 1685, pp. 1-15.
Singh et al., Rational Design of Small Peptides for Optimal Inhibition of Cyclooxygenase-2: Development of a Highly Effective Anti-Inflammatory Agent, Journal of Medicinal Chemistry, 59(8):3920-3934 (2016).
Sirohi et al., The 3.8 A resolution cryo-EM structure of Zika virus, Science, 352(6284):467-470 (2016).
Stettler et al., Specificity, cross-reactivity, and function of antibodies elicited by Zika virus infection, Science, 353(6301):823-826 (2016).
Stobiecka et al., Biosensors based on molecular beacons, Chemical Papers, 69:62-76 (2015).
Szybki. version 1.5.2. OpenEye Scientific Software, Santa Fe, NM. http://www.eyesogen.com.
Takahashi et al., Induction of CD8+ cytotoxic T cells by immunization with purified HIV-1 envelope protein in ISCOMs, Nature, 344:873-875 (1990).
Takeda et al., Construction of chimaeric processed immunoglobulin genes containing mouse variable and human constant region sequences, Nature, 314:452-454 (1985).
Tambunan et al., Designing disulfide cyclic peptide as fusion inhibitor that targets denv envelope proteine, Jurnal Teknologi, 78:4-3 (2016).
Tang et al., Quantum dot-DNA aptamer conjugates coupled with capillary electrophoresis: A universal strategy for ratiometric detection of organophosphorus pesticides, Talanta, 146:55-61 (2016).
Titus et al., Human T cells targeted with anti-T3 cross-linked to antitumor antibody prevent tumor growth in nude mice, J. Immunol., 138:4018-4022 (1987).
Todorovska et al., Design and application of diabodies, triabodies and tetrabodies for cancer targeting, Journal of Immunological Methods, 248:47-66, (2001).
Tomasevic et al., A high affinity recombinant antibody to the human EphA3 receptor with enhanced ADCC activity, Growth Factors, 32:223-235 (2014).
Trott et al., AutoDock Vina: Improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading, J. Computational Chem., 31(2):455-461 (2010).
Tsekenis et al., Label-less immunosensor assay for myelin basic protein based upon an ac impedance protocol, Analytical Chemistry, 80(6):2058-2062 (2008).
Vida. version 4.1.1. OpenEye Scientific Software, Santa Fe, NM. http://www.eyesogen.com.
Warren et al., Future prospects for vaccine adjuvants, CRC Critical Reviews in Immunology, 8:83-101 (1988).
Weaver et al., Zika virus: History, emergence, biology, and prospects for control, Antiviral Research, 130:69-80 (2016).
Acebes et al., Rational enzyme engineering through biophysical and biochemical modeling, ACS Catalysis, 6(3), 1624-1629 (2016).
Armour et al., Recombinant human IgG molecules lacking Fcgamma receptor I binding and monocyte triggering activities, Eur. J. Immunol., 29:2613-2624, (1999).
Barba-Spaeth et al., Structural basis of potent Zika-dengue virus antibody cross-neutralization, Nature, 536(7614):48-53 (2016).
Binz and Pluckthun, Engineered proteins as specific binding reagents, Curr Opin Biotechnol., 16(4):459-69 (2005).
Bohm et al., Combinatorial docking and combinatorial chemistry: design of potent non-peptide thrombin inhibitors, J. Computer-Aided Molecular Design, 13(1):51-56 (1999).
Bunker et al., Rational Design of Liposomal Drug Delivery Systems, a Review: Combined Experimental and Computational Studies of Lipid Membranes, Liposomes and Their PEGylation, Biochimica et Biophysica Acta (BBA)-Biomembranes, 1858(10):2334-2352 (2016).
Burch et al., Priming Tissue-specific Cellular Immunity in a Phase I Trial of Autologous Dendritic Cells for Prostate Cancer, Clinical Cancer Research, 6(6):2175-2182 (2000).
Burmeister et al., Crystal structure of the complex of rat neonatal Fc receptor with Fc, Nature, 372:379-383, (1994).
Byrne et al., A tale of two specificities: bispecific antibodies for therapeutic and diagnostic applications, Trends Biotechnol., 31:621-632 (2013).
Campos et al., Prolonged detection of Zika virus RNA in urine samples during the ongoing Zika virus epidemic in Brazil, Journal of Clinical Virology, 77:69-70 (2016).
Chan et al., Therapeutic antibodies for autoimmunity and inflammation, Nat Rev Immunol., 10:301-316 (2010).
Cole et al., Monoclonal Antibodies and Cancer Therapy, Alan R Liss Inc, New York N.Y., 77-96 (1985).
Cote et al., Generation of human monoclonal antibodies reactive with cellular antigens, Proc Natl Acad Sci., 80:2026-2030 (1983).
Cross et al., Analytical Validation of the ReEBOV Antigen Rapid Test for Point-of-Care Diagnosis of Ebola Virus Infection, The Journal of Infectious Diseases, 214(suppl3):S210-S217 (2016).
Cuesta et al., Multivalent antibodies: when design surpasses evolution, Trends in Biotechnology, 28:355-362 (2010).
Dai et al., Structures of the Zika Virus Envelope Protein and Its Complex with a Flavivirus Broadly Protective Antibody, Cell Host & Microbe., 19(5):696-704 (2016).
Davis et al., Single Chain Antibody (SCA) Encoding Genes: One-Step Construction and Expression in Eukaryotic Cells, Nature Biotechnology, 9:165-169 (1991).
Dudley et al., Adoptive cell therapy for patients with metastatic melanoma: evaluation of intensive myeloablative chemoradiation preparative regimens, J. Clin. Oneal., 26:5233-5239 (2008).
Friend et al., Phase I study of an engineered aglycosylated humanized CD3 antibody in renal transplant rejection, Transplantation, 68:1632-1637 (1999).
Gofflot et al., Immuno-quantitative polymerase chain reaction for detection and quantitation of prion protein, Journal of Immunoassay and Immunochemistry, 25(3):241-258 (2004).
Gold et al., Aptamers As Therapeutic And Diagnostic Agents, J. Biotechnol., 74:5-13 (2000).
Gong et al., Peptide Aptamer: a powerful potential tool in plant functional genomics (English Abstract only), Hereditas., 32(6):548-554 (2010).
Gonzalez-Diaz et al., Plasmonic Au/Co/Au nanosandwiches with Enhanced Magneto-Optical Activity, Small, 4(2):202-205 (2008).
Grant et al., A paper-based immunoassay to determine HPV vaccination status at the point-of-care, Vaccine, 34(46):5656-5663 (2016).
Gridelli et al., Efficient Human Fetal Liver Cell Isolation Protocol Based on Vascular Perfusion for Liver Cell-Based Therapy and Case Report on Cell Transplantation, Liver Transplantation, 18:226-237 (2012).
Haskard and Archer, The production of human monoclonal autoantibodies from patients with rheumatoid arthritis by the EBV-hybridoma technique, Journal of Immunological Methods, 74(2):361-367 (1984).
Hawkins et al., Conformer Generation with Omega: Algorithm and Validation Using High Quality Structures from the Protein Databank and Cambridge Structural Database, Journal of Chemical Information and Modeling, 50(4):572-584 (2010).
Hawkins et al., Conformer Generation with Omega: Learning from the Data Set and the Analysis of Failures, Journal of Chemical Information and Modeling, 52(11):2919-2936, (2012).
Heffron et al., Antibody responses to Zika virus proteins in pregnant and non-pregnant macaques, PLoS Neglected Tropical Diseases, 12(11):e0006903 (2018).
Holliger et al., Engineered antibody fragments and the rise of single domains, Nat. Biotechnol., 23:1126-1136 (2005).
Hoover et al., Adjuvant active specific immunotherapy for human colorectal cancer: 6.5-year median follow-up of a phase III prospectively randomized trial, J Clin Oncol., 11:390-399 (1993).
Hu et al., Minibody: A Novel Engineered Anti-Carcinoembryonic Antigen Antibody Fragment (Single-Chain Fv-CH3) Which Exhibits Rapid, High-Level Targeting of Xenografts, Cancer Research, 56:3055-3061 (1996).
Huse et al., Generation of a large combinatorial library of the immunoglobulin repertoire in phage lambda, Science, 246:1275-1281 (1989).
Huzly et al., High specificity of a novel Zika virus ELISA in European patients after exposure to different flaviviruses, Euro. Surveillance, 21:1-4 (2016).
Hwang et al., High sensitive and selective electrochemical biosensor: Label-free detection of human norovirus using affinity peptide as molecular binder, Biosensors and Bioelectronics, 87:164-170 (2017).
International Preliminary Report on Patentability for Corresponding International Application No. PCT/US2017/059128, dated May 9, 2019.
International Preliminary Report on Patentability for Corresponding International Application No. PCT/US2017/059129, dated May 9, 2019.
International Search Report and Written Opinion for Corresponding International Application No. PCT/US2017/059128, dated Feb. 16, 2018.
International Search Report and Written Opinion for Corresponding International Application No. PCT/US2017/059129, dated Jan. 18, 2018.
International Search Report and Written Opinion for corresponding International Application No. PCT/US2019/061862, dated Feb. 14, 2020.
Karpovsky et al., Production of target-specific effector cells using hetero-cross-linked aggregates containing anti-target cell and anti-Fc gamma receptor antibodies, J. Exp. Med., 160:1686-701 (1984).
Kelley et al., POSIT: flexible shape-guided docking for pose prediction, Journal of Chemical Information and Modeling, 55(8):1771-1780 (2015).
Khan et al., Human fetal liver-derived stem cell transplantation as supportive modality in the management of end-stage decompensated liver cirrhosis, Cell Transplant, 19:409-418 (2010).
Kim et al., Development of a novel peptide aptamer-based immunoassay to detect Zika virus in serum and urine, Theranostics, 8(13):3629-3642 (2018).
Koehler et al., Continuous cultures of fused cells secreting antibody of predefined specificity, Nature, 256:495-497, (1975).
Kortt et al., Dimeric and trimeric antibodies: high avidity scFvs for cancer targeting, Biomolecular Engineering, 18:95-108, (2001).
Kozbor et al., The production of monoclonal antibodies from human lymphocytes, Immunol. Today, 4:72-79 (1983).
Related Publications (1)
Number Date Country
20190385705 A1 Dec 2019 US
Provisional Applications (1)
Number Date Country
62684960 Jun 2018 US