Engineered human extracellular DNASE enzymes

Information

  • Patent Grant
  • 11352613
  • Patent Number
    11,352,613
  • Date Filed
    Monday, October 25, 2021
    2 years ago
  • Date Issued
    Tuesday, June 7, 2022
    a year ago
Abstract
The present disclosure provides a library of engineered DNASE proteins (including DNASE1, DNASE1-LIKE 1, DNASE1-LIKE 2, DNASE1-LIKE 3, DNASE2A, DNASE2B) that allows to select drug candidates for developing therapeutics for treating conditions characterized by neutrophil extracellular trap (NET) accumulation and/or release. In accordance with the invention, the selected DNase variant has improved properties, including properties amenable to clinical development, including manufacturing, toxicology, pharmacokinetic, and/or use in therapy.
Description
BACKGROUND

Inflammation is an essential host response to control invading microbes and heal damaged tissues. Uncontrolled and persistent inflammation causes tissue injury in a plethora of inflammatory disorders. Neutrophils are the predominant leukocytes in acute inflammation. During infections neutrophils generate neutrophil extracellular traps (NETs), lattices of DNA-filaments decorated with toxic histones and enzymes that immobilize and neutralize bacteria. However, excessive NET formation may harm host cells due to their cytotoxic, proinflammatory, and prothrombotic activity.


DNASE1 (D1) forms along with DNASE1-LIKE 1 (D1L1), DNASE1-LIKE 2 (D1L2) and DNASE1-LIKE 3 (D1L3), the DNASE1-protein family, a group of homologous secreted DNase enzymes. DNASE2A and DNASE2B form an additional group of homologous extracellular DNase enzymes. DNASE1- and DNASE2-protein family members are evolutionary conserved and expressed in various species, including humans. Recombinant human DNASE1- and DNASE2-protein family members provide drug candidates for NET-associated diseases, but the physical, enzymatic, toxicological, and pharmacokinetic properties of these enzymes are not ideal for clinical applications. Thus, there is a need for engineered DNase enzymes for use in therapy that have improved properties, including properties amenable to clinical development, including manufacturing, toxicology, pharmacokinetic (including protease resistance), and/or use in therapy.


SUMMARY

The present invention provides candidates of engineered human extracellular DNase proteins (e.g., variants of DNASE1 (D1), DNASE1-LIKE 1 (D1L1), DNASE1-LIKE 2 (D1L2), DNASE1-LIKE 3 Isoform 1 (D1L3), DNASE1-LIKE 3 Isoform 2 (D1L3-2), DNASE2A (D2A), and DNASE2B (D2B)) that are useful for treating conditions characterized by extracellular DNA, extracellular chromatin, and/or neutrophil extracellular trap (NET) accumulation and/or release. In accordance with aspects of the invention, DNase variants described herein are more amenable to clinical development, including manufacturing, toxicology, pharmacokinetic, and/or use in therapy.


In some aspects, the invention provides a method for making a DNase therapeutic composition for treating an extracellular chromatin or NET-associated disorder. The method comprises evaluating a plurality of extracellular DNase variants for one or more characteristics, including enzymatic activity, nucleic acid substrate preference, potential for recombinant expression in prokaryotic or eukaryotic host cells, immunogenic potential in humans and animals, and pharmacodynamics in animal models. An extracellular DNase variant is selected with the desired enzymatic, physical, and pharmacodynamics profile, and is formulated for administration to a patient, e.g., for either systemic or local administration.


In various embodiments, the DNase variant evaluated and selected for use in therapy in accordance with embodiments of the invention comprise an amino acid sequence that is at least 80% identical to the enzyme defined by any one of SEQ ID NOS: 1 to 7, with one or more building block substitutions or C-terminal modifications as described herein. In some embodiments, the DNase variant comprises an N-terminal or C-terminal fusion to a half-life extending moiety, such as albumin, transferrin, an Fc, or elastin-like protein.


In various embodiments, a selected DNase variant is formulated with a pharmaceutically acceptable carrier for systemic, local, or topical administration.


In other aspects, the invention provides a method for treating a subject in need of extracellular DNA degradation, extracellular chromatin degradation, extracellular trap (ET) degradation and/or neutrophil extracellular trap (NET) degradation. The method comprises administering a therapeutically effective amount of the extracellular DNase variant in accordance with the disclosure.


Other aspects and embodiments of the disclosure will be apparent from the following detailed description.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1 illustrates the approach for engineering DNase variants for therapeutic applications using Building Block Protein Engineering.



FIG. 2 shows an alignment of DNASE1-LIKE 3 Isoform 1 proteins from different species. Amino acids that are non-conserved in human DNASE1 are highlighted. Such non-conserved amino acids can be transferred to human DNASE1-LIKE 3 Isoform 1 for developing a variant for therapy. The DNASE1-LIKE 3 Isoform 1 proteins used for this analysis were Human DNASE1-LIKE 3 Isoform 1, UniProtKB: Q13609; NCBI Reference Sequence: NP_004935.1 (SEQ ID NO: 4); Pan troglodytes (Chimpanzee) DNase1L3 UniProtKB: A0A2I13RHL6 (and H2QMU7) (SEQ ID NO: 33); Papio anubis (Olive baboon) DNASE1L3, UniProtKB: A0A2I3NFJ3 (SEQ ID NO: 34); Mouse Dnase113, UniProtKB: 055070 (SEQ ID NO: 31); Rat DNase1L3, UniProtKB: 089107 (SEQ ID NO: 32); Oryctolagus cuniculus (Rabbit) DNase1L3, UniProtKB: G1SE62 (SEQ ID NO: 35); Canis lupus familiaris (Dog) DNase1L3, UniProtKB: F1P9C1 (SEQ ID NO: 36); Sus scrofa (Pig) DNase1L3, UniProtKB: A0A287B132 (SEQ ID NO: 37); Cavia porcellus (Guinea pig) DNase1L3, UniProtKB: A0A286XK50 (SEQ ID NO: 38); Bos taurus (Cow) DNase1L3, UniProtKB: F1MGQ1 (SEQ ID NO: 39); and Loxodonta africana (African elephant) DNase1L3, UniProtKB: G3SXX1 (SEQ ID NO: 40)



FIG. 3 shows an alignment of two members of the human DNASE1 proteins family, DNASE1-LIKE 1 and DNASE1-LIKE 3 Isoform 1. Amino acids that are conserved among human DNASE1-LIKE 1 (NCBI Reference Sequence: NP_006721.1; SEQ ID NO: 2) and DNASE1-LIKE 3 Isoform 1 (NCBI Reference Sequence: NP_004935.1; SEQ ID NO: 4) are highlighted. The non-conserved amino acids can be transferred from human DNASE1-LIKE 1 to DNASE1-LIKE 3 Isoform 1 or vice versa for developing variants for therapy, respectively.



FIG. 4 shows the concept of building block engineering of homologous proteins. The technology transfers single or multiple variable amino acids, which are flanked by conserved single or multiple variable amino acids, between a donor and recipient protein.



FIG. 5 shows an amino acid sequence alignment of DNase1 and DNase1L3 of mouse (SEQ ID NOs: 31 and 32), rat (SEQ ID NOs: 33 and 34), chimpanzee (SEQ ID NOs: 35 and 36), and human (SEQ ID NOs: 1 and 4). The N-terminal signal peptide, corresponding to N-terminal 22 amino acids of DNase1 is shown in light grey and conserved amino acids are highlighted in a darker shade of grey. Variable amino acids are not highlighted and serve as Building Blocks that can be transferred from DNase1 to DNase1L3 and vice versa. Abbreviations: AA, amino acid.



FIGS. 6A-FIG. 6B show lists of Building Blocks in human DNase1 (D1) and human DNase1L3 (D1L3). FIG. 6A shows amino acids that are conserved in D1 and D1L3, which serve as N- and C-anchors, respectively. Building blocks are variable amino acids in D1 and D1L3. Mutations that transfer Building Blocks from D1L3 to D1 are shown. FIG. 6B shows N- and C-anchors in D1L3. Mutations that transfer Building Blocks from D1 to D1L3 are listed. AA: amino acid.



FIG. 7 shows an application of the building block engineering of homologous proteins. The application uses as an initial screening step, the transfer of clusters of building blocks between a homologous donor and recipient protein. Additional optional steps are the transfer of individual building blocks, followed by the transfer of individual amino acids. In a final step (not shown), multiple amino acids, building blocks, and building block clusters may be combined to degenerate a chimeric enzyme.



FIG. 8 shows characterization of DNase1 variants (Dlv) featuring building blocks from DNase1L3 (D1L3). Zymography showed dsDNA degrading activity as dark circles. The dsDNA degrading activity correlates with the diameter. Samples without activity show the loading well as small black spot (e.g. Ctrl). Agarose gel electrophoresis (AGE) of DNA isolated from digested chromatin shows a shift from high-molecular weight DNA to lower or low-molecular weight DNA that correlates with chromatin degrading activity. Building block substitutions that cause an increase in chromatin degrading activity are highlighted in dark shade. Samples without such effect are shown in light shade. A DNase1 variant featuring the combination of building blocks 11, 12-14, 26, 41-48, and 49 shows similar chromatin degrading activity than wild-type DNase1L3.



FIG. 9 illustrates that the mutation Q282_S305delinsK in D1L3 Isoform 1 increases the activity to degrade high-molecular weight chromatin of DNASE1L3.



FIG. 10 shows the location of NLS2 sequence superimposed with a sequence alignment of C-termini of the S305del, K303_S305del, V294_S305del, K291_S305del, R285_S305del, and S283_S305de1 deletion mutants, which lack 1, 3, 12, 15, 21, and 23 C-terminal amino acids, respectively.



FIG. 11 shows a western blot of wild type D1L3 and BD-deleted D1L3 expressed in furin-overexpressing CHO cells. CHO cells without overexpression of furin were included as control. Data suggest that cleavage of the C-terminal tail or a portion thereof could act as an activation signal for D1L3.



FIG. 12A shows the expression vectors containing WT-DNASE1L3 or BDD-DNASE1L3 fused via a linker to Fc fragment, and used for expression in CHO cells. FIG. 12B shows SDS-PAGE protein gels of Fc fusion proteins purified via protein A/G from supernatants of CHO cells expressing WT-DNASE1L3 or BDD-DNASE1L3 fused via a linker to Fc fragment. Both gels show bands of approximately 65-70 kDa, which reflects the molecular weight of the full-length proteins. An additional lower molecular weight (between 29-44 kDa) is detected, when CHO cells express the WT-DNASE1L3 construct.



FIG. 13 shows a table of C-terminal amino acid sequences of DNASE1L3 variants that are secreted by cells expressing WT DNASE1L3 cDNA. The C-termini were specifically truncated at the three clusters of paired basic amino acids, resulting in deletions of 1, 8, 9, 13, 14 and 15 amino acids of the BD.





DETAILED DESCRIPTION

As used herein, the term “neutrophil extracellular trap” or “NET” refers to any extracellular trap (“ET”) comprising extracellular DNA formed by cells such as, but not limited to, neutrophils, monocytes, macrophages, basophils, eosinophils, mast cells, cancer cells, injured cells (e.g., injured endothelial cells), and the like. Unless the context indicates otherwise, the terms NET and ET are used interchangeably herein.


The similarity of nucleotide and amino acid sequences, i.e. the percentage of sequence identity, can be determined via sequence alignments as known in the art. Such alignments can be carried out with several art-known algorithms, such as with the mathematical algorithm of Karlin and Altschul (Karlin & Altschul (1993) Proc. Natl. Acad. Sci. USA 90: 5873-5877), with hmmalign (HMMER package, http://hmmer.wustl.edu/) or with the CLUSTAL algorithm (Thompson, J. D., Higgins, D. G. & Gibson, T. J. (1994) Nucleic Acids Res. 22, 4673-80). Exemplary algorithms are incorporated into the BLASTN and BLASTP programs of Altschul et al (1990) J. Mol. Biol. 215: 403-410. When utilizing BLAST programs, the default parameters of the respective programs are used.


In various aspects, the invention provides a protein engineering technology that is based on a transfer of a single amino acid or multiple-adjacent amino acids, termed “building block”, between two members of a protein family, such as DNase1 or DNase 2 protein family members to generate enzymatically active variants. A “building block” is defined by amino acids that are variable between two or more members of the DNase protein family. These variable amino acids are flanked by amino acids that are conserved between two or more members of the DNase-protein family (“anchors”). The variable single amino acid or multiple contiguous amino acids (“building blocks”) are exchanged between members of the DNase-protein family by implanting them between conserved single amino acid or multiple contiguous amino acids (“anchors”).


This approach is referred to herein as “building-block protein engineering.” Where three or more amino acids are transferred in a building block, up to ⅓ of the amino acids transferred may be further substituted. For example, where three to six amino acids are transferred as a building block, one or up to two resides may be further substituted. In some embodiments, four or more amino acids are transferred as a building block substitution, and up to 25% of the transferred amino acids are further substituted, e.g., with conservative or non-conservative amino acid modifications. For example, where four, eight, or twelve amino acids are transferred, one, two, or three amino acids (respectively) may be further substituted in the building block substitution.


The present invention provides candidates of engineered human extracellular DNase proteins (e.g., variants of DNASE1 (D1), DNASE1-LIKE 1 (D1L1), DNASE1-LIKE 2 (D1L2), DNASE1-LIKE 3 Isoform 1 (D1L3), DNASE1-LIKE 3 Isoform 2 (D1L3-2), DNASE2A (D2A), and DNASE2B (D2B)) that are useful for treating conditions characterized by extracellular DNA, extracellular chromatin, and/or neutrophil extracellular trap (NET) accumulation and/or release. In accordance with aspects of the invention, DNase variants described herein are more amenable to clinical development, including manufacturing, toxicology, pharmacokinetic, and/or use in therapy.


In some aspects, the invention provides a method for making a DNase therapeutic composition for treating a NET-associated disorder or disorder characterized by pathological accumulation of extracellular chromatin. The method comprises evaluating a plurality of extracellular DNase variants for one or more characteristics, including enzymatic activity, nucleic acid substrate preference, suitability for recombinant expression in prokaryotic or eukaryotic host cells, immunogenic potential in humans and animals, and pharmacodynamics in animal models. An extracellular DNase variant is selected with the desired enzymatic, physical, and pharmacodynamics profile, and is formulated for administration to a patient, e.g., for either systemic or local administration.


In various embodiments, at least 5 or at least 10, or at least 20, or at least 50 extracellular DNase variants are evaluated, with the variants selected from one or more of D1 variants, D1L1 variants, D1L2 variants, D1L3 isoform 1 variant, D1L3 isoform 2 variants, D2A variants, and D2B variants as described herein. As described herein, one or more (or all) variants may comprise at least one building block substitution, half-life extension moiety, and/or other mutation or variation described herein. In some embodiments, the method evaluates one or more D1L1 variants described herein. In some embodiments, the method evaluates one or more D1L2 variants described herein. In some embodiments, the method evaluates one or more D1L3 variants described herein. In some embodiments, the method evaluates one or more D1L3-2 variants described herein. In some embodiments, the method evaluates one or more D2A variants described herein. In some embodiments, the method evaluates one or more D2B variants described herein. In some embodiments, the method evaluates one or more D1 variants described herein.


In various embodiments, the invention provides a recombinant variant of human extracellular DNase enzymes comprising one or more amino acid alterations resulting in an altered pH and temperature optimum, requirement for divalent cations for enzymatic activity, mechanisms of enzymatic inhibition (e.g. salt, divalent cations, actin, heparin, proteases), substrate affinity and specificity (e.g. single-stranded DNA, double-stranded DNA, chromatin, NETs, plasmid DNA, mitochondrial DNA), localization upon secretion (e.g. membrane-bound, extracellular matrix), localization signals (e.g. nuclear localization signal, membrane anchor), glycosylation sites, disulfide-bonds and unpaired cysteines, compatibility with GMP-compliant in vitro expression systems (e.g. bacteria. yeast, mammalian cells), compatibility with carriers (e.g. PEGylation, Fc fragment, albumin), compatibility with GMP-compliant purification methods (e.g. anion exchange resins, cation exchange resins), toxicological profile, tissue penetration, pharmacokinetics and pharmacodynamics. In accordance with this disclosure, candidate DNase variants can be selected with desired properties for therapy.


In various embodiments, DNase variants will comprise at least one building block substitution, using a Building Block Protein Engineering technology. The Building Block Engineering approach is described in PCT/US2018/047084 (corresponding to WO 2019/036719 and U.S. Pat. No. 10,696,956), which are hereby incorporated by reference in its entirety. This approach involves providing a protein-protein alignment of donor and recipient DNase enzymes, and identifying variable amino acid sequences for transfer (“building block”). The variable amino acid(s) are flanked by one or more conserved amino acids in the donor and recipient DNase enzymes (upstream and downstream of the building block). These building blocks can be swapped between recipient and donor proteins, to produce a chimeric enzyme. The donor and recipient DNase enzymes can be selected from members of the DNASE1- or DNASE2-protein family. Accordingly, for example, human DNASE1 and human DNASE1L1 can be selected as donor and recipient DNase proteins, respectively. Alternatively, donor and recipient DNase can be selected from a DNase proteins that are expressed in different species. Accordingly, for example, bovine and human DNASE1 can be selected as donor and recipient DNase proteins, respectively.


As used herein, when referring to sequence identity with wild-type extracellular DNase enzymes, and unless stated otherwise, sequences refer to mature enzymes lacking the signal peptide. Further, unless stated otherwise, amino acid positions are numbered with respect to the full translated extracellular DNase sequence, including signal peptide, for clarity. Accordingly, for example, reference to sequence identity to the enzyme of SEQ ID NO: 1 (human D1) refers to a percent identity with the mature enzyme having L23 at the N-terminus, reference to sequence identity to the enzyme of SEQ ID NO: 2 (human D1L1) refers to a percent identity with the mature enzyme having F19 at the N-terminus, reference to sequence identity to the enzyme of SEQ ID NO: 3 (human D1L2) refers to a percent identity with the mature enzyme having L22 at the N-terminus, reference to sequence identity to the enzyme of SEQ ID NO: 4 (human D1L3) refers to a percent identity with the mature enzyme having M21 at the N-terminus, reference to sequence identity to the enzyme of SEQ ID NO: 5 (human D1L3-2) refers to a percent identity with the mature enzyme having M21 at the N-terminus, reference to sequence identity to the enzyme of SEQ ID NO: 6 (human D2A) refers to a percent identity with the mature enzyme having C19 at the N-terminus, and reference to sequence identity to the enzyme of SEQ ID NO: 7 (human D2B) refers to a percent identity with the mature enzyme having A28 at the N-terminus.


The term “delins” refers to a deletion between two indicated amino acids, with an insertion of an amino acid or sequence of amino acids at the site of the deletion. For example, the notation E91_P92delinsSR means that the amino acids from E91 to P92 are deleted and the amino acids SR are inserted at the site of the deletion (e.g., the resulting amino acid sequence will have S91 and R92).


The term “ins” refers to an insertion of amino acids between two indicated amino acids. For example, the notation E91_P92insSR means that the amino acids SR are inserted between E91 and P92, resulting in the sequence E91, S92, R93, and P94.


The term “del” refers to a deletion of one amino acid or two and more amino acids between two indicated amino acids. For example, the notation E91del means that the amino acid E91 is deleted, whereas the notion E91_P93del means that the three amino acids from E91 and P93 are deleted.


The engineered variants of human extracellular DNase enzymes may comprise one or more additional amino acid substitutions, additions (insertions), deletions, or truncations in the amino acid sequence of the human enzyme (SEQ ID NO: 1 to 7). Amino acid substitutions may include conservative and/or non-conservative substitutions. For example, “conservative substitutions” may be made, for instance, on the basis of similarity in polarity, charge, size, solubility, hydrophobicity, hydrophilicity, and/or the amphipathic nature of the amino acid residues involved. The 20 naturally occurring amino acids can be grouped into the following six standard amino acid groups: (1) hydrophobic: Met, Ala, Val, Leu, Ile; (2) neutral hydrophilic: Cys, Ser, Thr; Asn, Gln; (3) acidic: Asp, Glu; (4) basic: His, Lys, Arg; (5) residues that influence chain orientation: Gly, Pro; and (6) aromatic: Trp, Tyr, Phe. “Conservative substitutions” are defined as exchanges of an amino acid by another amino acid listed within the same group of the six standard amino acid groups shown above. For example, the exchange of Asp by Glu retains one negative charge in the so modified polypeptide. In addition, glycine and proline may be substituted for one another based on their ability to disrupt α-helices. As used herein, “non-conservative substitutions” are defined as exchanges of an amino acid by another amino acid listed in a different group of the six standard amino acid groups (1) to (6) shown above.


In various embodiments, the DNASE1 (D1) variant evaluated and selected for use in therapy in accordance with embodiments of the invention comprises an amino acid sequence that is at least 80% identical to the enzyme defined by SEQ ID NO: 1, with one or more building block substitutions.


In some aspects, the building block substitutions are selected from non-human D1 proteins and result in variants of human D1, which feature one or more of the following mutations: K24R, I25M, Q31R, T32S, E35D, V44S, V44T, V44A, S45V, S45K, S45N, S45H, Q49K, Q49R, S52Q, S52R, R53L, I56V, A57V, L58V, V59I, S60T, T68V, D75N, N76E, N76K, N76T, N76E, N76Y, N76S, Q79R, Q79E, D80K, D80H, A81K, A81I, A81D, P82A, P82T, D83N, D83G, T84N, T84A, Y85F, H86R, Y87F, Y87H, V88I, V89I, V89A, N96K, N96R, N96S, S97T, R101Q, V105L, Y106F, D109S, Q110R, Q110K, A113V, A113I, S114L, S116T, Y108Q, Y108H, Y108L, P125S, N128T, T130S, N132S, N132A, A1365, I137V, R139K, F141S, F141H, S142C, R143P, R143H, F144Y, F144S, F144L, V147K, V147Q, R148Q, R148S, E149K, I152V, P154A, A1575, G160E, G160T, G160L, G160S, D161E, V163A, S164S, D167N, A168S, D175N, Q177W, Q177R, E178Q, E178K, E178H, G181D, G181H, E183Q, E183N, V185I, M186V, L187F, G194D, C195Y, R199T, R199A, R199S, P200S, P200A, P200T, P200L, Q202H, S204A, W209R, T210M, T210E, P212S, T213A, T213I, T213P, Q215K, Q215R, P219L, S221T, S221N, A226V, A226S, T227S, T227K, P228S, H230N, A232P, M241T, M241A, M241P, M241S, R244Q, G245D, G245A, G245H, G245R, G245S, D250N, D250S, D250E, D250G, L253V, L253A, L253M, N256D, A259V, A260E, Y261F, G262R, S264T, D265N, D265S, D265E, Q266E, L267M, L267T, Q269E, Q269L, M280T, M280A, K282R, K282A, K282T, K282insK, and K282insR.


In some embodiments, the building block substitutions to D1 are selected from human D1L1 and result in variants of human D1 which feature one or more of the following mutations: M1 G3del, K5G8delinsHYPT, A12F, L14Q15delinsAN, V21_K24delinsQAFR, A26C, I30A, T32_T36delinsRLTLA, M38_I47delinsVAREQVMDTL, Q49R, S52A, Y54C, A57_V59delinsMVL, R63V, H66_T68delinsSGS, V70_K72delinsIPL, D75_N76delinsRE, Q79delinsRF, A81_T84delinsGSGP, H86_V89delinsSTLS, E91_P92delinsPQ, N96_S97delinsST, K99M, R101T, L103_V105delinsVYF, P108_D115delinsSHKTQVLS, Y118V, D120N, G122_N128delinsED, T130V, A136_R139delinsFVAQ, F141_I152delinsSLPSNVLPSLVL, A157_A158delinsTT, G160_A164delinsKAVEK, I166_D167delinsLN, Y173F, D175E, G177_E179delinsQSK, M186L, G194D, S196_R207delinsASLTKKRLDKLE, W209R, S211E, T213G, Q215H, L217V, P219A, S221_A222delinsGE, A226_P228delinsVRAS, A232T, I236V, V238 A239delinsLH, M241_L243delinsERC, G245_S251delinsSLLHT, L253_P254delinsAA, N256D, Q259_G262delinsPTSQG, S264_L267delinsTEEE, Q269_A270delinsLN, M280E, and K282delinsKLSQAHSVQPLSLTVLLLLSLLSPQLCPAA.


In certain embodiments, the building block substitutions to D1 are selected from human D1L2 and result in variants of human D1 which feature one or more of the following mutations: R2G, M4 KSdelinsPRA, G8A, L11W, A14E, L16Q18delinsA, A20_S22delinsTAA, K24R, A26G, T32S, E35 T36delinsDS, M38V, N40_V44delinsDPACG, Y46I, V48_Q49delinsAK, S52_R53delinsAG, I56L, S65_H66delinsPD, T68S, S70_A71delinsGK, L74_L77delinsMEQI, Q79_T84delinsSVSEHE, H86_Y87delinsSF, V89S, E91Q, D95_Q96delinsNS, R101M, P108K, Q110A, A113V, S116T, Y118L, P119D, G122_N128delinsPE, T130V, N132S, A136_I137delinsFV, R139K, F141_F144delinsSAPG, E146_V153delinsGERAPPLPSRRALTPPLPAAAQNLVLI, G160_D161delinsHQ, Q177_E178delinsID, L182_E182delinsTD, V185_M188delinsMLFL, G194D, P200_Q202delinsAQD, S204_S205delinsAA, W209_T210delinsRS, P212_T213delinsEV, Q230K, A226_H230delinsVGNSD, V239_A240delinsAC, M241_L242delinsAR, G245_V248delinsRSLK, D250Q, L253_N256delinsTVHD, A259_Y262delinsEEF, S264_L267delinsDQTQ, Q269L, Y275F, M280T, and K282insFHR.


In some embodiments, the building block substitutions to D1 are selected from human DNASE1-LIKE 3 Isoform 1 (D1L3) and result in variants of human D1, which feature one or more of the following mutations: M1_L6delinsMSRE, G8_A9deinsAP, A12L, A14_G19delinsLLSIHS, V21_K24delinsLAMR, A26_A27delinsCS, I30_T32delinsVRS, S36T, M38_Y46delinsQEDKNAMDV, Q49_S52delinsKVIK, Y54C, A57I, Q60M, V62_R63delinsIK, H66_K72delinsNNRICPI, L74_N76delinsMEK, Q79_D83delinsRNSRRGI, H86N, V89I, E91_P92delinsSR, S97T, R101Q, L103A, V105L, R107 Q110delinsKEKL, A113_D115delinsVKR, Y118H, D120H, G122_N128delinsYQDGDA, T130V, N132S; A136_I137delinsFV, R139W, F141Q, R143_F144delinsPH, E146A, R148_E149delinsKD, A151V, V153I, A157_A 158delinsTT, G160_A162delinsETS, A164K, A168E, Y170_D171delinsVE, L174T, Q177_K179delinsKHR, G181_L182delinsKA, D184_L187delisNFIF, R199_Q202delisPKKA, S204_S205delinsKN, W209R, S211D, T213R, Q215V, P219G, S221_A222delinsQE, A226_P228delinsVKKS, H230N, V238_A239delinsLR, M241_A246delinsQEIVSS, D250K, A252_P254delinsNSV, N256D, A259K, G262K, S264_L267delinsTEEE, Q269_I271delinsLDV, Y275F, V279_M280delinsFK, and K282delinsQSSRAFTNSKKSVTLRKKTKSKRS.


In some embodiments, the building block substitutions to D1 are selected from human DNASE1-LIKE 3 Isoform 2 (D1L3-2) and result in variants of human D1, which feature one or more of the following mutations: M1_L6delinsMSRE, G8_A9deinsAP, A12L, A14_G19delinsLLSIHS, V21_K24delinsLAMR, A26_A27delinsCS, I30_T32delinsVRS, T36S, M38_Y46delinsQEDKNAMDV, Q49_S52delinsKVIK, Y52C, A57I, Q60M, V62_R63delinsIK, H66_K72delinsNNRICPI, L74N76delinsMEK, Q79_Y106deL, P108_Q110delinsEKL, A113_D11SdelinsVKR, Y118H, D120H, G122_N128delinsYQDGDA, T130V, N132S, A136_I137delinsFV, R139W, F141Q, R143_F144delinsPH, E146A, R148_E149delinsKD, A151V, VI531, A157_A158delinsAA, G160_A162delinsETS, A164K, Y170D171delinsVE, L174T, Q177_K179delinsKHR, G181_L 182delinsKA, D184_L 187delisNFIF, R199_Q202delisPKKA, S204_S205delinsKN, W209R, S211D, T231R, Q215V, P219G, S221_S222delinsQE, A226_P228delinsVKKS, H230N, V238_A239delinsLR, M241 _A246delinsQEIVSS, D250K, A252_P254delinsNSV, N256D, G262K. S264_L267delinsTEEE, Q269_I271delinsLDV, Y275F, V279_M280delinsFK, and K282delinsQSSRAFTNSKKSVTLRKKTKSKRS.


In various embodiments, the D1 variant evaluated in accordance with the disclosure comprises the D1 wildtype amino acid sequence or a variant sequence described herein, fused to the C-terminus of a carrier protein, with a linking amino acid sequence. In some embodiments, the carrier protein is Fc fragment or albumin. In some embodiments, the carrier protein is human albumin. The linking sequence can be a flexible linker predominately composed of Gly, or Gy and Ser. In some embodiments, the linker is composed of Gly and amino acids having hydrophilic side chains (e.g., Ser, Thr). In some embodiments, the linker is from 5 to 20 amino acids. An exemplary linker has the structure (GGGGS)3.


In various embodiments, the peptide linker may be a flexible linker, a rigid linker, or in some embodiments a physiologically-cleavable linker (e.g., a protease-cleavable linker). In some embodiments, the linker is 5 to 100 amino acids in length, or is 5 to 50 amino acids in length.


Linkers, where present, can be selected from flexible, rigid, and cleavable peptide linkers. Flexible linkers are predominately or entirely composed of small, non-polar or polar residues such as Gly, Ser and Thr. An exemplary flexible linker comprises (GlyySer)n linkers, where y is from 1 to 10 (e.g., from 1 to 5), and n is from 1 to about 10, and in some embodiments, is from 3 to about 6. In exemplary embodiments, y is from 2 to 4, and n is from 3 to 8. Due to their flexibility, these linkers are unstructured. More rigid linkers include polyproline or poly Pro-Ala motifs and α-helical linkers. An exemplary α-helical linker is A(EAAAK)nA, where n is as defined above (e.g., from 1 to 10, or 2 to 6). Generally, linkers can be predominately composed of amino acids selected from Gly, Ser, Thr, Ala, and Pro. Exemplary linker sequences contain at least 10 amino acids, and may be in the range of 15 to 35 amino acids. Exemplary linker designs are provided as SEQ ID NOS: 41 to 51.


In some embodiments, the variant comprises a linker, wherein the amino acid sequence of the linker is predominately glycine and serine residues, or consists essentially of glycine and serine residues. In some embodiments, the ratio of Ser and Gly in the linker is, respectively, from about 1:1 to about 1:10, from about 1:2 to about 1:6, or about 1:4. Exemplary linker sequences comprise S(GGS)4GSS (SEQ ID NO: 46), S(GGS)9GS (SEQ ID NO: 47), (GGS)9GS (SEQ ID NO: 48). In some embodiments, the linker has at least 10 amino acids, or at least 15 amino acids, or at least 20 amino acids, or at least 25 amino acids. For example, the linker may have a length of from 15 to 30 amino acids. In various embodiments, longer linkers of at least 15 amino acids can provide improvements in yield upon expression in Pichia pastoris.


In other embodiments, the linker is a physiologically-cleavable linker, such as a protease-cleavable linker. For example, the protease may be a coagulation pathway protease, such as activated Factor XII. In certain embodiments, the linker comprises the amino acid sequence of Factor XI (SEQ ID NO: 49) and/or prekallikrein (SEQ ID NO: 50 or 51) or a physiologically cleavable fragment thereof. In other embodiments, the linker includes a peptide sequence that is targeted for cleavage by a neutrophil specific protease, such as neutrophil elastase, cathepsin G, and proteinase 3.


In various embodiments, the human DNASE1-LIKE 1 (DIL1) variant evaluated and selected for use in therapy in accordance with embodiments of the invention comprise an amino acid sequence that is at least 80% identical to the enzyme defined by SEQ ID NO: 2, with one or more building block substitutions.


In some embodiments, the building block substitutions to D1L1 are selected from non-human D1L1 proteins and result in variants of human D1L1 which feature one or more of the following mutations: A26T, Q27H, A32T, A325, V34L, A35T, A35I, R36K, Q38S, Q38E, Q38H, Q38Y, Q38P, Q38D, M40K, M40L, T42I, L43F, R45Q, R45K, L47V, M53T, S61A, S62T, G63Q, G63D, G63N, G63S, S64N, S64A, S64K, S64T, A65T, P66L, P66S, L68F, R71Q, R71E, E72K, N74S, R75K, F76Y, D77K, D77Q, D77Y, D77G, G78A, G78S, G78N, G78D, G80R, G80K, P81S, P81F, P81C, S83R, T84F, T84S, L85H, S86N, S86K, P88S, P88D, Q89L, Q89M, S93N, S93G, T94A, M96V, M96K, T98K, V100A, F102I, H106D, K107R, K107E, K107R, T108A, Q109E, V110L, L111R, S112N, S112D, S112E, S113F, V115Q, V115L, V115M, N117D, N117E, N117P, N177S, E119T, E119Q, E119K, V122I, V122L, A124T, A130G, A130C, Q131H, Q131W, S133T, L134F, P135R, N137D, N137K, V138T, V138I, L142V, V143A, A153D, K156P, K156N, K156T, L159K, Y162H, D163E, D163T, E167D, V168A, S169Y, S169A, Q170R, Q170G, H171R, S174N, S174T, K175E, K175Q, S176N, V177M, V177I, A188T, T191A, T191N, D196K, D196N, D196S, D196A, D196G, E199L, E199A, E199V, E203K, E203D, E203Q, P204A, P204V, P204T, H207R, H207S, V209A, I210V, A211P, E214D, E241V, H223N, T225A, V229I, L231V, L231M, E234Q, E234V, R235G, R235T, R235L, C236L, R237Q, S238M, S238K, S238G, L240M, H241K, H241Q, H241S, H241R, T242A, T242S, T242N, T242G, A244T, D247N, T240K, T250R, Q250Q, S251T, S251R, Q252R, Q252G, T255N, T255S, E258Q, E259Q, N261R, N261K, M261T, I262V, E271D, K273S, K273N, K273D, K273A, L274del, S275Q, S275K, S275R, Q276A, A277T, A277V, H278P, H278Q, 5279G, S279N, S279R, C279S, I280V, A280V, Q281P, Q281L, L283H, L283P, S284Y, S284C, S284H, 5284G, T286A, T286S, T286V, V287T, V287A, F287F, G287V, L288A, L288S, L289S, L289V, L289M, S292L, S292P, S295P, S295T, S295A, P296S, Q297E, L298C, C299D, C299G, C299S, P300L, A301Q, A301V, and A302M.


In some embodiments, the building block substitutions to D1L1 are selected from human D1 and result in variants of human D1L1 which feature one or more of the following mutations: M1delinsMRGM, H2_T5delinsKLLG, F9A, A13_N14delinsLQ, Q17_R20delinsVLSK, C22A, A26I, R28_A32delinsTFGET, V34_L43delinsMSNATLVSYI, R45Q, A48S, C50Y, M53_L55delinsALV, V59R, S62_S64delisHLT, 166_L68delinsVGK, R71_E72delinsDN, R75_F76delinsQ, G78_P81delinsAPDT, S83_S86delinsHYVV, P88_Q89delinsEP, S93_T94delinsNS, M96K, T98R, V100_F102delinsLFV, S105_S112delinsPDQVSAVD, V115Y, N117D, E119_D120delinsGCEPCGN, V122T, F128_Q131 delinsAIVR, S133_L144delinsFSRFTEVREFAI, T149_T150delinsAA, K152_K156delinsGDAVA, L158_N159delinsID, F165Y, E167D, Q173_K175delinsGLE, M188L, D186G, A188_E199delinsSYVRPSQWSSIR, R201W, E203S, G205T, H207Q, V209L, A211P, G213_E214delinsSA, V218_S221delinsATP, T225A, V229I, L231_H232delinsVA, E234_C236delinsMLL, S238_T242delinsGAVVPDS, A244_A245delinsLP, D247N, P249_Q253delinsQAAYG, T255_E258delinsSDQL, L260_N261delinsQA, E271M, and L274_A302del.


In some embodiments, the building block substitutions to D1L1 are selected from human D1L2 and result in variants of human D1L1 which feature one or more of the following mutations: H2_Y3delinsGG, TSR, F9_L12delinsWALE, N14A, A16_Q17delinsTA, F19L, C22G, A26I, R28_A32delinsSFGDS, A35_R45delinsSDPACGSIIAK, R49_50CdelinsGY, 152_L55delinsLALV, V59R, S61_G63delinsPDL, I66_L68delinsVSA, L70_L73delinsMEQI, R75_P81delinsSVSEHE, T84_L85delinsFV, P88_Q89delinsQP, S93_T94delinsDQ, M96K, T98M, V100_F102delinsLFV, S105_Q109delinsKDAVS, L111_S113delinsVDT, V115L, N117P, E119_P120delinsPE, A124S, A130_Q131delinsVK, L134A, S136_V138delinsGTGERAPP, S141_L142insRRALTPPPLPAAAQN, V145I, T149_T150delinsAA, K152_K156delinsHQAVA, L158_N159delinsID, F165Y, E167D, S169_H171 delinsIDK, V177_L179delinsMLF, A188_E199delinsSYVRAQDWAAIR, T202_G205delinsSSEV, H207K, V209L, A211P, G213_E214delinsSA, R219_H223delinsGNSD, T225A, V229I, L231_H232delinsAC, E234A, C236L, S238_L239delinsRS, H241_T242delinsKPQS, A244_F246delinsTVH, P249_S251delinsQEE, Q253G, T255 E258delinsDQTQ, N261A, Y266F, E271T, and L274_A302delinsFHR.


In some embodiments, the building block substitutions to D1L1 are selected from human DNASE1-LIKE 3 Isoform I (DIL3) and result in variants of human D1L1, which feature one or more of the following mutations: H2_T5delinsSREL, L7_L8insPLL, F9L, I11_G14delinsLSIHS, Q17L, F19M, A23S, A26_A32delinsVRSFGES, V34_V39delinsQEDKNA, T42_L43delinsVI, R45_A48delinsKVIK, M53_Q56delinsILVM, V58_V59delinsIK, S62_I66delinsNNRIC, L68I, L70_E72delinsMEK, F76_S79delinsNSRR, G80_P81delinsIT, S83_S86delinsNYVI, P88_Q89delinsPQ, S92N, M96K, T98Q, V100_F102delinsAFL, R104_Q109delinsKEKLVS, L111_S112delinsKR, V115H, N117H, E119_D120delinsYQDGDA, A124S, A130_Q131delinsVW, S133_L134delinsQS, S136L142delinsHTAVKDF, L144_V145delinsII, K152_E155delinsETSV, L158_A160delinsIDE, Y162_D163delinsVE, F165_E167delinsVTD, S169_H171delinsKHR, Q173_V177delinsKAENF, D186G, A188_T191delinsSYVP, R194_E199delinsAWKNIR, E203D, G205R, H207V, V209L, A211G, G213Q, R219_A220delinsKK, H223N, T225A, V229I, H232R, E234_R237delinsQEIV, L239_A245delinsSVVPKSNSV, P249_Q253delinsQKAYK, N261_I262delinsDV, Y266F, V270E271delinsFK, K273_L274delinsQS, Q276R, H278_L283delinsFTNSKK, L285V, and V287_A302delinsLRKKTKSKRS.


In some embodiments, the building block substitutions to D1L1 are selected from human DNASE1-LIKE 3 Isoform 2 (D1L3-2) and result in variants of human D1L1, which feature one or more of the following mutations: H2 T5delinsSREL, A7delinsPLL, F9L, I11_G14delinsLSIHS, Q17L, F19M, S23A, A26_A32delinsVRSFGES, V34_V39delinsQEDKNA, T42_L43delinsVI, R45_A48delinsKVIK, M53_Q56delinsILVM, V58_V59delinsIK, S62_I66delinsNNRIC, L68I, L70_E72delinsMEK, F75_Q103del, S105_Q109delinsEKLVS, L111_S112delinsKR, V115H, N117H, E119_D120delinsYQDGDA, A124S, A130_Q131delinsVW, S133_L134delinsQS, S136_L142delinsHTAVKDF, L144_V145delinsII, K152_E155delinsETSV, L158_A160delinsIDE, Y162_D163delinsVE, F165_E167delinsVTD, S169_H171 delinsKHR, Q173_V177delinsKAENF, D186G, A188_T191delinsSYVP, R194_E199delinsAWKNIR, E203D, G205R, H207V, V209L, A211G, G213Q, R219_A220delinsKK, H223N, T225A, V229I, H232R, E234_R237delinsQEIV, L239_A245delinsSVVPKSNSV, P249_Q253delinsQKAYK, N261_I262delinsDV, Y266F, V270_E271delinsFK, K273_L274delinsQS, Q276R, H278_L283delinsFTNSKK, L285V, and V287_A302delinsLRKKTKSKRS.


In certain embodiments, the D1L1 protein variant contains one or more amino acid substitutions, additions, or deletions in the C-terminal tail domain defined by SEQ ID NO: 8. The C-terminal tail domain or a portion thereof may be deleted. In some embodiments, at least 3, 5, 8, 10, 12, 15, 18, or 23 amino acids of the C-terminal tail domain are deleted.


In various embodiments, the D1L1 variant evaluated in accordance with the disclosure comprises the D1L1 wildtype amino acid sequence or a variant sequence described herein, fused to the C-terminus of a carrier protein, with a linking amino acid sequence. In some embodiments, the carrier protein is Fc fragment or albumin. In some embodiments, the carrier protein is human albumin. The linking sequence, which is also herein referred to as a peptide linker, or a linker, can be a flexible linker predominately composed of Gly, or Gy and Ser. In some embodiments, the linker is composed of Gly and amino acids having hydrophilic side chains (e.g., Ser, Thr). In some embodiments, the linker is from 5 to 20 amino acids. An exemplary linker has the structure (GGGGS)3.


The peptide linker may be a flexible linker, a rigid linker, or in some embodiments a physiologically-cleavable linker (e.g., a protease-cleavable linker). In some embodiments, the linker is 5 to 100 amino acids in length, or is 5 to 50 amino acids in length.


Linkers, where present, can be selected from flexible, rigid, and cleavable peptide linkers. Flexible linkers are predominately or entirely composed of small, non-polar or polar residues such as Gly, Ser and Thr. An exemplary flexible linker comprises (GlyySer)n linkers, where y is from 1 to 10 (e.g., from 1 to 5), and n is from 1 to about 10, and in some embodiments, is from 3 to about 6. In exemplary embodiments, y is from 2 to 4, and n is from 3 to 8. Due to their flexibility, these linkers are unstructured. More rigid linkers include polyproline or poly Pro-Ala motifs and α-helical linkers. An exemplary α-helical linker is A(EAAAK)nA, where n is as defined above (e.g., from 1 to 10, or 2 to 6). Generally, linkers can be predominately composed of amino acids selected from Gly, Ser, Thr, Ala, and Pro. Exemplary linker sequences contain at least 10 amino acids, and may be in the range of 15 to 35 amino acids. Exemplary linker designs are provided as SEQ ID NOS: 41 to 51.


In some embodiments, the variant comprises a linker, wherein the amino acid sequence of the linker is predominately glycine and serine residues, or consists essentially of glycine and serine residues. In some embodiments, the ratio of Ser and Gly in the linker is, respectively, from about 1:1 to about 1:10, from about 1:2 to about 1:6, or about 1:4. Exemplary linker sequences comprise S(GGS)4GSS (SEQ ID NO: 46), S(GGS)9GS (SEQ ID NO: 47), (GGS)9GS(SEQ ID NO: 48). In some embodiments, the linker has at least 10 amino acids, or at least 15 amino acids, or at least 20 amino acids, or at least 25 amino acids. For example, the linker may have a length of from 15 to 30 amino acids. In various embodiments, longer linkers of at least 15 amino acids can provide improvements in yield upon expression in Pichia pastoris.


In other embodiments, the linker is a physiologically-cleavable linker, such as a protease-cleavable linker. For example, the protease may be a coagulation pathway protease, such as activated Factor XII. In certain embodiments, the linker comprises the amino acid sequence of Factor XI (SEQ ID NO: 49) and/or prekallikrein (SEQ ID NO: 50 or 51) or a physiologically cleavable fragment thereof. In other embodiments, the linker includes a peptide sequence that is targeted for cleavage by a neutrophil specific protease, such as neutrophil elastase, cathepsin G, and proteinase 3.


In various embodiments, the human DNASE1-LIKE 1 (D1L2) variant evaluated and selected for therapy in accordance with this disclosure comprises an amino acid sequence that is at least 80% identical to the enzyme defined by SEQ ID NO: 3, with one or more building block substitutions.


In some embodiments, the building block substitutions to D1L2 are selected from non-human D1L2 proteins and result in variants of human D1L2, which feature one or more of the following mutations: L22K, I24V, I29V, S35N, S35H, S35R, 535T, V37A, S38L, A41D, A41V, A41G, G43I, S44G, S44i, I45V, K48Q, L55I, L55V, A56T, A56M, P64A, S70D, S70T, A71T, A71L, A71S, A71V, M73L, E74Q, N77H, S78R, E81K, E81R, E83N, S85G, S85N, Q90E, Q90K, Q96H, F103Y, V104I, K107D, A109V, A109T, A109K, V110A, V113L, V113M, D1145, D114E, L117Q, P119S, E122G, V124A, V124F, S126N, E128D, F134V, A136V, A136T, G1385, G138R, T139S, T139C, S148C, A151P, P154A, A159P, A160G, A161P, A161T, Q162D, Q162K, Q162R, Q162T, N163K, N163E, L164V, L164F, I167V, H174N, Q175H, A178T, A178V, D192N, G195N, T196S, D198V, M199L, M199I, S210K, R213K, Q215H, A218P, A219S, E226Q, V227I, S243T, A252V, C253S, A255S, A255V, R256H, L257M, R259K, S260T, L261V, Q264H, T267S, T267A, D270N, G276D, G276S, T280S, T280D, T280A, A284C, I286V, L295F, F297S, F297T, F297P, H298R, and R299del.


In some embodiments, the building block substitutions to D1L2 are selected from human D1 and result in variants of human D1L2, which feature one or more of the following mutations: G2R, P4_A6delinsMK, A9G, W12L, EISA, A16_G18delinsLLQ, T19_A21delinsAVS, R23K, G25A, S31T, D34_S35delinsET, V37M, D39_G43delinsNATLV, I45Y, A47_K48delinsVQ, A51_G52delinsSR, L55I, P64_D65delinsSH, S67T, S70_A71delinsGK, M73_I76delinsLDNL, S78_E83delinsQDAPDT, S85_F86delinsHY, S88V, Q90E, D95_Q96delinsNS, M100R, K107P, A109Q, V112A, T115S, L117Y, P119D, P121_E122delinsGCEPCGN, V124T, S126N, F130_V131delinsI, K133R, S135_G138delinsFSRF, G140I167delinsEVREFAIV, H174_Q175delinsGD, I191_D192delinsQE, T196_D197delinsLE, M199_L202delinsVMLM, D208G, A214_D216delinsPSQ, A218_A219delinsSS, R223_S224delinsWT, E226_V227delinsPT, K229Q, V240_D244delinsATPTH, A252_C253delinsVA, A225_R256delinsML, R259_K262delinsGAVV, Q264D, T267 D270delinsLPFN, E273_E275delinsAAY, D278_Q281delinsSDQL, L283Q, F283Y, T294M, and F297_R299del.


In some embodiments, the building block substitutions to D1L2 are selected from human D1L1 and result in variants of human D1L2, which feature one or more of the following mutations: G2_G3delinsHY, R5T, W12_E15delinsFLIL, A17N, T19_A20delinsAQ, L22F, G25C, I29A, S31_S35delinsRLTLA, S38_K48delinsAREQVMDTLVR, G52_Y53delinsRC, L55_V58delinsIMVL, R62V, P64_L66delinsSSG, V69_A71delinsIPL, M73_I76delinsLREL, S78_E83delinsRFDGSGP, F86_V87delinsTL, Q90_P91delinsPQ, D95_Q96delinsST, K98M, M100T, L102_V104delinsVYF, K107_S111delinsSHKTQ, V113_T115delinsLSS, L117V, P119N, P121_E122delinsED, S126A, V132_K133delinsAQ, A136L, G138_P145delinsSNV, R149_N163del, I167V, A171_A172delinsTT, H174_A178delinsKAVEK, I180_D181delinsLN, Y187F, D189E, I191_K193delinsSQH, M199_F201delinsVIL, S210_R221delinsASLTKKRLDKLE, S224_V227delinsTEPG, K229H, L231V, P233A, S235_A235delinsGE, G241_D244delinsRASTH, A246T, 1250V, A252_C253delinsLH, A255E, L257C, R259_S260delinsSL, K262_S265delinsHT, T267_H269delinsAAF, Q272_E274delinsPTS, G276Q, D278_Q281delinsTEEE, A284N, F289Y, T294E, and F297_R299delinsLSQAHSVQPLSTVLLLLSLLSPQLCPAA.


In some embodiments, the building block substitutions to D1L2 are selected from human DNASE1-LIKE 3 Isoform 1 (D1L3) and result in variants of human D1L2, which feature one or more of the following mutations: G2_R5delinsSREL, L7P, A9_A10delinsL, W12_A13delinsLL, S15_A20delinsSIHSAL, L22M, G25_A26delinsCS, I29_Q30delinsVR, D34E, V37_S38delinsQE, P40_I45delinsKNAMDV, A47V, I49_Y53delinsVIKRS, L55_A56delinsII, Q59M, V61_R62delinsIK, P64_A71delinsSNNRICPI, Q75_I76delinsKL, N77_S78insRS, V79_E83delinsRRGIT, S85_F86delinsNY, S88I, Q90_P91delinsSR, D95_Q96delinsNT, M100Q, L102A, V104L, R106_A109delinsKEKL, V113_T115delinsKRS, L117H, P119H, P121_E122delinsYQDGDA, K133W, S135_A136delinsQS, G138H, G140_A159del, A161_L164delinsVKDF, L166I, A171_A172delinsTT, H174_A176delinsETS, A178K, A182E, Y184_D185delinsVE, L188T, 1191_K193delinsKHR, G195_L200delinsKAENFI, L202M, D208G, R213_D126delinsPKKA, A218_A129delinsKN, S224_V227delinsTDPR, K229V, P233G, S235_A236delinsQE, G241_N242delinsKK, S243_D244insTN, A252_C253delinsLR, A255_R259delinsQEIVS, L261_K262delinsVV, Q264K, A266_T267delinsNS, H269F, E273_G276delinsKAYK, D278_Q281delinsTEEE, A284_I285delinDV, V293_T294delinsFK, K296_H298delinsQSS, and R295insAFTNSKKSVTLRKKTKSKRS.


In some embodiments, the building block substitutions to D1L2 are selected from human DNASE1-LIKE 3 Isoform 2 (D1L3-2) and result in variants of human D1L2, which feature one or more of the following mutations: G2 R5delinsSREL, L7P, A9 _A10delinsL, W12_A13delinsLL, S15_A20delinsSIHSAL, L22M, G25_A26delinsCS, I29_Q30delinsVR, D34E, V37_S38delinsQE, P40_I45delinsKNAMDV, A47V, I49_Y53delinsVIKRS, L55_A56delinsII, Q59M, V61_R62delinsIK, P64_A71delinsSNNRICPI, Q75_I76delinsKL, K107_A109delinsEKL, V113_T115delinsKRS, L117H, P119H, P121_E122delinsYQDGDA, K133W, S135_A136delinsQS, G138H, G140_A159del, A161_L164delinsVKDF, L166I, A171_A172delinsTT, H174_A176delinsETS, A178K, A182E, Y184_D185delinsVE, L188T, 1191_K193delinsKHR, G195_L200delinsKAENFI, L202M, D208G, R213_D126delinsPKKA, A218_A129delinsKN, S224_V227delinsTDPR, K229V, P233G, S235_A236delinsQE, G241_N242delinsKK, S243_D244insTN, A252_C253delinsLR, A255_R259delinsQEIVS, L261_K262delinsVV, Q264K, A266_T267delinsNS, H269F, E273_G276delinsKAYK, D278_Q281delinsTEEE, A284_I285delinDV, V293_T294delinsFK, K296_H298delinsQSS, and R295insAFTNSKKSVTLRKKTKSKRS.


In certain embodiments, the D1L2 protein variant contains one or more amino acid substitutions, additions, or deletions in the proline-rich extension domain defined by SEQ ID NO: 9. The proline-rich extension domain or a portion thereof may be deleted, including a deletion (or truncation) of at least 3 amino acids, at least 5 amino acids, or at least 10 amino acids.


In various embodiments, the D1L2 variant evaluated in accordance with the disclosure comprises the D1L2 wildtype amino acid sequence or a variant sequence described herein, fused to the C-terminus of a carrier protein, with a linking amino acid sequence. In some embodiments, the carrier protein is Fc fragment or albumin. In some embodiments, the carrier protein is human albumin. The linking sequence can be a flexible linker predominately composed of Gly, or Gy and Ser. In some embodiments, the linker is composed of Gly and amino acids having hydrophilic side chains (e.g., Ser, Thr). In some embodiments, the linker is from 5 to 20 amino acids. An exemplary linker has the structure (GGGGS)3.


The peptide linker may be a flexible linker, a rigid linker, or in some embodiments a physiologically-cleavable linker (e.g., a protease-cleavable linker). In some embodiments, the linker is 5 to 100 amino acids in length, or is 5 to 50 amino acids in length.


Linkers, where present, can be selected from flexible, rigid, and cleavable peptide linkers. Flexible linkers are predominately or entirely composed of small, non-polar or polar residues such as Gly, Ser and Thr. An exemplary flexible linker comprises (GlyySer)n linkers, where y is from 1 to 10 (e.g., from 1 to 5), and n is from 1 to about 10, and in some embodiments, is from 3 to about 6. In exemplary embodiments, y is from 2 to 4, and n is from 3 to 8. Due to their flexibility, these linkers are unstructured. More rigid linkers include polyproline or poly Pro-Ala motifs and α-helical linkers. An exemplary α-helical linker is A(EAAAK)nA, where n is as defined above (e.g., from 1 to 10, or 2 to 6). Generally, linkers can be predominately composed of amino acids selected from Gly, Ser, Thr, Ala, and Pro. Exemplary linker sequences contain at least 10 amino acids, and may be in the range of 15 to 35 amino acids. Exemplary linker designs are provided as SEQ ID NOS: 41 to 51.


In some embodiments, the variant comprises a linker, wherein the amino acid sequence of the linker is predominately glycine and serine residues, or consists essentially of glycine and serine residues. In some embodiments, the ratio of Ser and Gly in the linker is, respectively, from about 1:1 to about 1:10, from about 1:2 to about 1:6, or about 1:4. Exemplary linker sequences comprise S(GGS)4GSS (SEQ ID NO: 46), S(GGS)9GS (SEQ ID NO: 47), (GGS)9GS (SEQ ID NO: 48). In some embodiments, the linker has at least 10 amino acids, or at least 15 amino acids, or at least 20 amino acids, or at least 25 amino acids. For example, the linker may have a length of from 15 to 30 amino acids. In various embodiments, longer linkers of at least 15 amino acids can provide improvements in yield upon expression in Pichia pastoris.


In other embodiments, the linker is a physiologically-cleavable linker, such as a protease-cleavable linker. For example, the protease may be a coagulation pathway protease, such as activated Factor XII. In certain embodiments, the linker comprises the amino acid sequence of Factor XI (SEQ ID NO: 49) and/or prekallikrein (SEQ ID NO: 50 or 51) or a physiologically cleavable fragment thereof. In other embodiments, the linker includes a peptide sequence that is targeted for cleavage by a neutrophil specific protease, such as neutrophil elastase, cathepsin G, and proteinase 3.


In some embodiments, the human DNASE1-LIKE 3 Isoform 1 (D1L3) variant evaluated and selected for use in therapy in accordance with embodiments of the invention may comprise an amino acid sequence that is at least 80% identical to the enzyme defined by SEQ ID NO: 4, and may have one or more building block substitutions. As further disclosed in U.S. Pat. No. 10,696,956 (which is incorporated herein by reference), the D1L3 variant may comprise an amino acid sequence that has at least 90%, or at least 95%, or at least 97%, or at least 98% sequence identity with with the DNase enzyme defined by SEQ ID NO: 4.


In some embodiments, the building block substitutions to D1L3 are selected from non-human D1L3 proteins, such as chimpanzee, olive baboon, mouse, rat, dog, pig, guinea pig, cow, and elephant (See FIG. 2). In various embodiments, the building block substitution resultd in variants of human D1L3 which feature one or more of the following mutations: M21L, K22R, I22L, I22V, E33A, E33Y, E33G, S34A, S34T, Q36K, Q36R, E37A, E37Q, D48N, K39Q, K39H, K39R, K39C, N40E, N40Q, N40K, A41V, V44I, V53I, I53L, I54M, V57L, I60V, N64S, H64S, R66N, R66M, I70V, I70M, I70T, M72L, E73K, K74R, R77G, R81K, G82S, I83V, I83T, T84M, T84K, S91P, T91V, T91A, L105V, K107M, V111L, S112T, R115T, R115A, R115K, R115D, R115Q, S116K, S116N, S116Y, H118L, H118V, Y119F, H120G, Y122N, Q123E, D124A, D124S, D124N, G125E, A127V, A127T, V129A, F135Y, V137T, Q140H, S141A, H143F, H143Y, V146A, I152V, T157S, T160A, V162I, K163R, V169A, E170D, T173M, T173L, V175M, K176R, K176Q, H177S, H177R, R178Q, K180E, K180N, K181T, K181V, E183A, E183Q, A201S, K203Q, K203R, R212K, R212N, R212G, R212M, V214I, G218K, G218A, Q220E, Q220D, K227R, K2275, K227E, N239K, N239S, N239H, R239C, Q241P, E242D, E242N, V244I, S245N, S245R, K250R, K250D, K250R, K250G, K250N, K250Q, N252S, S253G, S253L, V254T, V254I, D256N, Q258R, Y261F, K262D, K262E, K262L, K262R, K262Q, T264S, E266S, E267K, E267Q, E267K, D270N, D270E, V271I, S282E, R285T, F287I, S290N, K291R, V294I, T295S, T295Q, L296V, L296P, L296S, R297K, K299R, T300K, T300A, S302G, S302A, S302V, S302T, K303N, K303S, K303R, R304H, R304S, S305P, S305T, and S305A. In some embodiments, the C-terminal tail is a sequence from mouse or rat D1L3 (See FIG. 2).


In some embodiments, the building block substitutions to D1L3 are selected from human D1 and result in variants of human D1L3 which feature one or more of the following mutations: M1_E4delinsMRGMKL, A6_P7delinsGA, L10A, L12_S17delinsAALLQG, L19_R22delinsVSLK, C24_S25delinsAA, V28_S30delinsIQT, S34T, Q36_V44delinsMSNATLVSY, K47_K50delinsQILS, C52Y, I55A, M58Q, I60_K61delinsVR, N64_I70delinsHLTAVGK, M72_K74delinsLDN, R77_I83delinsQDAPD, N86H, I89V, S91_R92delinsEP, T97S, Q101R, A103L, L105V, K107_L110delinsRPDQ, V113_R115delinsAVD, H118Y, H120D, Y122_A127delinsGCEPCGN, V129T, S131N, F135_V136delinsAI, W138R, Q140F, P142_H143delinsRF, A145E, K147_D148delinsRE, V150A, I152V, T156_T157delinsAA, E159_S161delinsGDA, K163A, E167A, V169_E170delinsYD, T173L, K176_R178delinsQEK, K180_A181delinsGL, N183_F186delinsDVML, P198_A201delinsRPSQ, K203_N204delinsSS, R208W, D210S, R212T, V214Q, G218P, Q220_E221delinsSA, V225_S228delinsATP, N230H, L238_R239delinsVA, Q241_S246delinsMLLRGA, K250D, N252_V254delinsALP, D256N, K259A, K262G, T264_E267delinsSDQL, L269_V271delinsQAI, F275Y, F279_K280delinsVM, and Q282_S205delinsK.


In some embodiments, the building block substitutions to D1L3 are selected from human D1L1 and result in variants of human D1L3 which feature one or more of the following mutations: S2_LSdelinsHYPT, P7_L9delinsL, L11F, L13_S17delinsILANG, L19delinsQ, M21F, S25A, V28_S34delinsAQRLTLA, Q36_A41delinsVAREQV, V44_I45delinsTL, K47_K50delinsRILA, 155_M58delinsMVLQ, I60_K61delinsVV, N6_C68delinsSGSAI, 170L, M72_L74delinsLRE, N78_R81delinsFDGS, I83_T84delinsP, N86_I89delinsSTLS, S91_R92delinsPQ, N96S, K99M, Q101T, A103_L105delinsVYF, K107_S112delinsRSHKTQ, K114_R115delinsLS, H118V, H120N, Y122_A127delinsED, S131A, V137_W138delinsAQ, Q140_S141delinsSL, H143_F149delinsSNVLPSL, I151_I152delinsLV, E159_V162delinsKAVE, I165_A167delinsLNA, V169_E170delinsYD, Y172_D174delinsFLE, K176_R178delinsSQH, K180_F184delinsQSKDV, G193D, S195_P198delinsASLT, A201_R206delinsRLDKLE, D210E, R212G, V214H, L216V, G218A, Q220G, K226_K227delinsRA, N230H, A232T, I236V, R239H, Q246_V249delinsERCR, S246_V254delinsLLHTAAA, Q258_K262delinsPTSFQ, D270_V271delinsNI, F275Y, F279_K280delinsVE, Q282_S283delinsKL, R285Q, F287_K292delinsHSVQPL, V294L, and L296_S205delinsVLLLLSLLSPQLCPAA.


In some embodiments, the building block substitutions to D1L3 are selected from human D1L2 and result in variants of human D1L3 which feature one or more of the following mutations: S2_L5delinsGGPR, P7L, L9delinsAA, L11_L12delinsWA, S14_L19delinsEAAGTA, M21L, C24_S25delinsGA, V28_R29delinsIQ, E33D, Q36_E37delinsVS, K39_V44delinsPACGSI, A47V, V48_C52delinsILAGY, I54_I55delinsLA, M58Q, 160_K61delinsVR, S63_I70delinsPDLSAVSA, K74_L75delinsQI, XX (deletion in Donor), R80_T84delinsVSEHE, N86_Y87delinsSF, 189S, S91_R92delinsQP, N96_T97delinsDQ, Q101M, A103L, L105V, K107_L110delinsRKDA, K114_S116delinsVDT, H118L, H120P, Y122_A127delinsPE, W138K, Q140_S141delinsSA, H143G, T144_A145insGERAPPLPSRRALTPPPLPA, V146_F149delinsAQNL, I151L, T156_T157delinsAA, E159_S161delinsHQA, K163A, E162A, V169_E170delinsYD, T173L, K176_R178delinsIDK, K180_I185delinsGTDDML, M187L, G193D, P198_A201delinsRAQD, K203_N204delinsAA, T209_R212delinsSSEV, V214K, G218P, Q220_E221delinsSA, K226_K227delinsGN, T229_N230delinsD, L238_R239delinsAC, Q241_S245delinsARLRR, V247_V248delinsLK, K250Q, N252_S253delinsAT, F255H, K259_K262delinsEEFG, T264_E267delinsDQTQ, D270_V271delinsAI, F279_K280delinsVT, Q282_S284delinsKFH, and A286_S305del.


In certain embodiments, the D1L3 protein variant contains one or more amino acid substitutions, additions, or deletions in the C-terminal tail amino acid sequence defined by SEQ ID NO: 10. The C-terminal tail domain or a portion thereof may be deleted. In some embodiments, at least 3, 5, 8, 10, 12, 15, 18, or 23 amino acids of the C-terminal tail domain are deleted. In some embodiments, such amino acid substitutions and/or deletions in the C-terminal tail (including building block substitutions of the C-terminal tail as disclosed above) reduce proteolysis in the C-terminal tail. In some embodiments, such substitutions can be made according to non-human D1L3 variants as described above, which may avoid one or more paired basic amino acid sites that are subject to protease cleavage.


In certain embodiments, the D1L3 protein variant contains one or more, e.g., 1, 2, 3, 4, 5, or more amino acid substitutions, additions, or deletions in the internal sequence defined by SEQ ID NO: 11 (which is absent from isoform 2), and which is optionally deleted in whole or in part.


In various embodiments, the D1L3 variant evaluated in accordance with the disclosure comprises the D1L3 wildtype amino acid sequence or a variant sequence described herein, fused to the C-terminus of a carrier protein, with a linking amino acid sequence. In some embodiments, the carrier protein is Fc fragment or albumin. In some embodiments, the carrier protein is human albumin. The linking sequence can be a flexible linker predominately composed of Gly, or Gy and Ser. In some embodiments, the linker is composed of Gly and amino acids having hydrophilic side chains (e.g., Ser, Thr). In some embodiments, the linker is from 5 to 20 amino acids. An exemplary linker has the structure (GGGGS)3.


The peptide linker may be a flexible linker, a rigid linker, or in some embodiments a physiologically-cleavable linker (e.g., a protease-cleavable linker). In some embodiments, the linker is 5 to 100 amino acids in length, or is 5 to 50 amino acids in length.


Linkers, where present, can be selected from flexible, rigid, and cleavable peptide linkers. Flexible linkers are predominately or entirely composed of small, non-polar or polar residues such as Gly, Ser and Thr. An exemplary flexible linker comprises (GlyySer)n linkers, where y is from 1 to 10 (e.g., from 1 to 5), and n is from 1 to about 10, and in some embodiments, is from 3 to about 6. In exemplary embodiments, y is from 2 to 4, and n is from 3 to 8. Due to their flexibility, these linkers are unstructured. More rigid linkers include polyproline or poly Pro-Ala motifs and α-helical linkers. An exemplary α-helical linker is A(EAAAK)nA, where n is as defined above (e.g., from 1 to 10, or 2 to 6). Generally, linkers can be predominately composed of amino acids selected from Gly, Ser, Thr, Ala, and Pro. Exemplary linker sequences contain at least 10 amino acids, and may be in the range of 15 to 35 amino acids. Exemplary linker designs are provided as SEQ ID NOS: 41 to 51.


In some embodiments, the variant comprises a linker, wherein the amino acid sequence of the linker is predominately glycine and serine residues, or consists essentially of glycine and serine residues. In some embodiments, the ratio of Ser and Gly in the linker is, respectively, from about 1:1 to about 1:10, from about 1:2 to about 1:6, or about 1:4. Exemplary linker sequences comprise S(GGS)4GSS (SEQ ID NO: 46), S(GGS)9GS (SEQ ID NO: 47), (GGS)9GS (SEQ ID NO: 48). In some embodiments, the linker has at least 10 amino acids, or at least 15 amino acids, or at least 20 amino acids, or at least 25 amino acids. For example, the linker may have a length of from 15 to 30 amino acids. In various embodiments, longer linkers of at least 15 amino acids can provide improvements in yield upon expression in Pichia pastoris. Further, longer linker sequences showed improved chromatin-degrading activity, as compared to shorter linker sequences.


In other embodiments, the linker is a physiologically-cleavable linker, such as a protease-cleavable linker. For example, the protease may be a coagulation pathway protease, such as activated Factor XII. In certain embodiments, the linker comprises the amino acid sequence of Factor XI (SEQ ID NO: 49) and/or prekallikrein (SEQ ID NO: 50 or 51) or a physiologically cleavable fragment thereof. In other embodiments, the linker includes a peptide sequence that is targeted for cleavage by a neutrophil specific protease, such as neutrophil elastase, cathepsin G, and proteinase 3.


In some embodiments, the DNASE1-LIKE 3 Isoform 2 (D1L3-2) variant evaluated and selected for use in therapy in accordance with embodiments of the invention comprise an amino acid sequence that is at least 80% identical to the enzyme defined by SEQ ID NO: 5, with one or more building block substitutions.


In some embodiments, the building block substitutions to D1L3-2 are selected from non-human D1L3 proteins and result in variants of human D1L3-2 which feature one or more of the following mutations: M21L, K22R, I22L, I22V, E33A, E33Y, E33G, S34A, S34T, Q36K, Q36R, E37A, E37Q, D48N, K39Q, K39H, K39R, K39C, N40E, N40Q, N40K, A41V, V44I, V53I, I53L, I54M, V57L, I60V, N64S, H64S, R66N, R66M, I70V, I70M, I70T, M72L, E73K, K74R, R77G, V81L, S82T, R85T, R85A, R85K, R85D, R85Q, S86K, S86N, S86Y, H88L, H88V, Y89F, H90G, Y92N, Q93E, D94A, D94S, D94N, G95E, A97V, A97T, V99A, F105Y, V107T, Q110H, S111A, H113F, H113Y, V116A, I122V, T127S, T130A, V132I, K133R, V139A, E140D, T143M, T143L, V145M, K146R, K146Q, H147S, H147R, R148Q, K150E, K150N, K151T, K151V, E153A, E153Q, A171S, K173Q, K173R, R182K, R182N, R182G, R182M, V184I, G188K, G188A, Q190E, Q190D, K197R, K197S, K197E, N209K, N209S, N209H, R209C, Q211P, E212D, E212N, V214I, S215N, S215R, K220R, K220D, K220R, K220G, K220N, K220Q, N222S, S223G, S223L, V224T, V224I, D226N, Q228R, Y231F, K232D, K232E, K232L, K232R, K232Q, T234S, E236S, E237K, E237Q, E237K, D240N, D240E, V241I, S252E, R255T, F257I, S260N, K261R, V264I, T265S, T265Q, L266V, L266P, L266S, R267K, K269R, T270K, T270A, S272G, S272A, S272V, S272T, K273N, K273S, K273R, R274H, R274S, S275P, S275T, and S275A.


In some embodiments, the building block substitutions to D1L3-2 are selected from human D1 and result in variants of human D1L3-2 which feature one or more of the following mutations: M1_E4delinsMRGMKLL, A6_P7delinsGA, L10A, L12_S17delinsAALLQG, L19_R22delinsVSLK, C24_S25delinsAA, V28_S30delinsIQT, S34T, Q36_V44delinsMSNATLVSY, K152_K156delinsGDAVA, C52Y, I55A, M58Q, I60_K61delinsVR, N64_I70delinsHLTAVGK, M72_K74delinsLDN, N76_R77in5QDAPDTYHYVVSEPLGRNSYKERYLFVY, E78_L80delinsPDQ, V83_R85delinsAVD, H88Y, H90D, Y92_A97delinsGCEPCGN, V99T, S101N, F105_V106delinsAI, W108R, Q110F, P112_H113delinsRF, A115E, K117_D118delinsRE, V120A, I122V, T126_T127delinsAA, E129_T131delinsGDA, K133A, V139_E140delinsYD, T143L, K146_R148delinsQEK, K150_A151delinsGL, N153_F156delinsDVML, P168_A171delinsRPSQ, K173_N1174delinsSS, R178W, D180S, R182T, V184Q, G188P, Q190_E191delinsSA, V195_S199delinsATP, N200H, L208_R209delinsVA, Q211_S216delinsMLLRGA, K220D, N222_V224delinsALP, D226N, K232G, T234_E237delinsSDQL, L239_V241delinsQAI, F245Y, F249_K250delinsVM, and Q252_S275delinsK.


In some embodiments, the building block substitutions to D1L3-2 are selected from human D1L1 and result in variants of human D1L3-2 which feature one or more of the following mutations: S2_L5delinsHYPT, P7_L9delinsA, L11F, L13_S17delinsILANG, L19delinsQ, M21F, S25A, V28_S34delinsAQRLTLA, Q36_A41delinsVAREQV, V44_I45delinsTL, K47_K50delinsRILA, 155_M58delinsMVLQ, I60_K61delinsVV, N6_C68delinsSGSAI, 170L, M72_L74delinsLRE, N76_R77insRFDGSGPYSTLSSPQLGRSTYMETYVYFYRSHKTQ, E78_S82delinsSHKTQ, K84_R85delinsLS, H88V, H90N, Y92_A97delinsED, S101A, V107_W108delinsAQ, Q110_S111delinsSL, H113_F119delinsSNVLPSL, I121_I122delinsLV, E129_V132delinsKAVE, I135_A137delinsLNA, V139_E140delinsYD, Y142_D144delinsFLE, K146_R148delinsSQH, K150_F154delinsQSKDV, G163D, S165_P168delinsASLT, A171_R176delinsRLDKLE, D180E, R182G, V184H, L186V, G188A, Q190G, K196_K227delinsRA, N200H, A192T, I206V, R209H, Q216_V219delinsERCR, S216_V224delinsLLHTAAA, Q228_K232delinsPTSFQ, D240_V241delinsN, F245Y, F249_K250delinsVE, Q252_S253delinsKL, R255Q, F257_K262delinsHSVQPL, V264L, and L266_S275delinsVLLLLSLLSPQLCPAA.


In some embodiments, the building block substitutions to D1L3-2 are selected from human D1L2 and result in variants of human D1L3-2 which feature one or more of the following mutations: S2_L5delinsGGPR, P7L, L9delinsAA, LL11_L12delinsWA, S14_L19delinsEAAGTA, M21L, C24_S25delinsGA, V28_R29delinsIQ, E33D, Q36_E37delinsVS, K39_V44delinsPACGSI, A47V, V48_C52delinsILAGY, I54_I55delinsLA, M58Q, I60_K61delinsVR, S63_I70delinsPDLSAVSA, K74_L75delinsQI, E78_L80delinsKDA, K84_S86delinsVDT, H88L, H90P, Y92_A97delinsPE, W108K, Q110_S111delinsSA, H113G, T114_A115insGERAPPLPSRRALTPPPLPA, V116_F119delinsAQNL, I121L, T126_T127delinsAA, E129_S141delinsHQA, K133A, E132A, V139_E140delinsYD, T143L, K146_R148delinsIDK, K150_I155delinsGTDDML, M157L, G163D, P168_A171delinsRAQD, K173_N174delinsAA, T179_R182delinsSSEV, V184K, G188P, Q190_E191delinsSA, K196_K197delinsGN, T199_N200delinsD, L208_R209delinsAC, Q211_S215delinsARLRR, V217_V218delinsLK, K220Q, N222_S223delinsAT, F225H, K229_K242delinsEEFG, T234_E237delinsDQTQ, D240_V241delinsAI, F249_K250delinsVT, and Q252_S254delinsKFH, A256_S275del.


In certain embodiments, the D1L3-2 protein variant contains one or more amino acid substitutions, additions, or deletions in the C-terminal tail domain defined by SEQ ID NO: 11. The C-terminal tail domain or a portion thereof may be deleted. In some embodiments, at least 3, 5, 8, 10, 12, 15, 18, or 23 amino acids of the C-terminal tail domain are deleted.


In various embodiments, the D1L3-2 variant evaluated in accordance with the disclosure comprises the D1L3-2 wildtype amino acid sequence or a variant sequence described herein, fused to the C-terminus of a carrier protein, with a linking amino acid sequence. In some embodiments, the carrier protein is Fc fragment or albumin. In some embodiments, the carrier protein is human albumin. The linking sequence can be a flexible linker predominately composed of Gly, or Gy and Ser. In some embodiments, the linker is composed of Gly and amino acids having hydrophilic side chains (e.g., Ser, Thr). In some embodiments, the linker is from 5 to 20 amino acids. An exemplary linker has the structure (GGGGS)3.


The peptide linker may be a flexible linker, a rigid linker, or in some embodiments a physiologically-cleavable linker (e.g., a protease-cleavable linker). In some embodiments, the linker is 5 to 100 amino acids in length, or is 5 to 50 amino acids in length.


Linkers, where present, can be selected from flexible, rigid, and cleavable peptide linkers. Flexible linkers are predominately or entirely composed of small, non-polar or polar residues such as Gly, Ser and Thr. An exemplary flexible linker comprises (GlyySer)n linkers, where y is from 1 to 10 (e.g., from 1 to 5), and n is from 1 to about 10, and in some embodiments, is from 3 to about 6. In exemplary embodiments, y is from 2 to 4, and n is from 3 to 8. Due to their flexibility, these linkers are unstructured. More rigid linkers include polyproline or poly Pro-Ala motifs and α-helical linkers. An exemplary α-helical linker is A(EAAAK)nA, where n is as defined above (e.g., from 1 to 10, or 2 to 6). Generally, linkers can be predominately composed of amino acids selected from Gly, Ser, Thr, Ala, and Pro. Exemplary linker sequences contain at least 10 amino acids, and may be in the range of 15 to 35 amino acids. Exemplary linker designs are provided as SEQ ID NOS: 41 to 51.


In some embodiments, the variant comprises a linker, wherein the amino acid sequence of the linker is predominately glycine and serine residues, or consists essentially of glycine and serine residues. In some embodiments, the ratio of Ser and Gly in the linker is, respectively, from about 1:1 to about 1:10, from about 1:2 to about 1:6, or about 1:4. Exemplary linker sequences comprise S(GGS)4GSS (SEQ ID NO: 46), S(GGS)9GS (SEQ ID NO: 47), (GGS)9GS (SEQ ID NO: 48). In some embodiments, the linker has at least 10 amino acids, or at least 15 amino acids, or at least 20 amino acids, or at least 25 amino acids. For example, the linker may have a length of from 15 to 30 amino acids. In various embodiments, longer linkers of at least 15 amino acids can provide improvements in yield upon expression in Pichia pastoris, and may provide for improved chromatin degrading activity.


In other embodiments, the linker is a physiologically-cleavable linker, such as a protease-cleavable linker. For example, the protease may be a coagulation pathway protease, such as activated Factor XII. In certain embodiments, the linker comprises the amino acid sequence of Factor XI (SEQ ID NO: 49) and/or prekallikrein (SEQ ID NO: 50 or 51) or a physiologically cleavable fragment thereof. In other embodiments, the linker includes a peptide sequence that is targeted for cleavage by a neutrophil specific protease, such as neutrophil elastase, cathepsin G, and proteinase 3.


In various embodiments, the DNASE2A (D2A) variant evaluated and selected for use in therapy in accordance with embodiments of the invention comprise an amino acid sequence that is at least 80% identical to the enzyme defined by SEQ ID NO: 6, with one or more building block substitutions.


In some embodiments, the building block substitutions to DNASE2A are selected from non-human D2A proteins and result in variants of human D2A, which feature one or more of the following mutations: Q25R, L38H, L38N, R39S, R39T, G40S, G42R, E43D, A44T, A44K, A44V, A45P, A45T, R47K, R47N, R47S, Q50T, Q50M, Q50R, L54M, L54F, E56Q, S57N, S57H, S57E, G59D, G59E, G60D, R62Q, R62S, R65V, R65A, A66G, L67Y, L67H, L67F, L67S, N69D, P71S, P71K, P71T, E72D, E72T, V75L, R77L, Q80L, R84Q, S85K, S85N, T87S, T87N, L93V, Q101K, P102S, P102Y, S103R, K104S, K104E, K104G, A105S, Q106R, Q106K, D107H, S109T, M110G, M110S, M110N, R111H, H122Q, D123E, V129I, N134R, P137S, P138R, A139S, A142G, A143V, S145T, H148P, S149N, S149G, C151Q, C151R, T152K, Y153F, L158I, F162L, F164L, A165T, A165S, S168A, S168P, S168L, K169R, K169G, K169D, K169N, M170I, G171S, K172R, W180L, W180M, N183D, Y184H, Q185K, Q185R, I180F, I180D, Q192R, E193K, F194L, D196Y, N199T, N199E, V201I, V201T, G203N, G203Q, S207L, S207R, Q208H, Q208R, E209G, I215V, T216I, Q220R, Q220K, A221K, A223T, V224T, V224S, F231C, S232G, K233N, A244S, A245E, T249S, N250T, H257Q, H257P, T259S, V260P, V260S, V260A, D269G, 1270A, 1270T, 1270V, W271Y, W271H, W271Q, Q272K, Q272H, V273I, L274F, N275D, N277T, Q278E, I279I, A280G, A285S, G286R, P287L, S288T, S288A, S288N, N290S, S291A, S301A, S301T, K303Q, K303E, G304R, I307A, I307V, Q316K, G317A, G317R, E319T, Q320H, L326V, A328T, L330V, L330M, A332S, L333F, Q338R, Q338K, P339S, N343D, N343A, Y344W, Y344C, Q345K, and Q345E.


In some embodiments, the building block substitutions to D2A are selected from human DNASE2B (D2B) and result in variants of human D2A, which feature one or more of the following mutations: 12 L6delinsKQKMM, A8R, C11_P13delinsRTSFALLFLGLFGVLG, G15 T18delinsATIS, Y20 S23delinsRNEE, Q25_P26delinsKA, V31_V32delinsTF, A37_L38delinsK, G40_G42delinsQNK, A44_R47delinsSGET, Q50E, K52L, E56_G60delinsSTIRS, D62_A66delinsKSEQ, I68M, S70_V75delinsDTKSVL, S78T, P81Q, R84delinsEAYA, N86_L90delinsKSNNT, F92Y, L94I, Q108_P109delinsGV, Q101K, S103_D107delinsVNK, V117L, L120_G124delinsWNRVQ, V129I, V132I, N134Q, P136del, A139_A143delinsIPEEG, S145_W146delinsDY, H148_Y153delinsPTGRRN, T156_L158delinsSGI, V160_S161delinsIT, P163_A165delinsKYN, F167_K172delinsYEAIDS, T175_Y178delinsLVCN, W180N, N183_I89delinsSCSIPAT, A192H, F194_V194delinsLIHMPQLCTRASS, Q208_E209delinsEI, W211_I215delinsGRLLT, T218Q, Q220_A221delnsAQ, A223_V224delinsQK, Q226_S227delinsLH, F231_K233delinsSDS, G235L, L238_G241delinsIFAA, L243M, A245_A246delinsQR, G248K, N250H, Q252_F255delinsLTET, H257_I262delinsQRKRQE, D269_Q272delinsLPYH, L274Y, Y276_Q278delinsIKA, A280_S288delinsKLSRHSY, N290S, T292_E293delinsYQ, S296A, V300I, P302Q, P305delinsTKNR, V309I, M312L, N315_Q320deinsSPHQAF, G322S, T325_L326delinsFI, L330_K335delinsNWQIYQ, P339G, K342_N343delinsLY, Q345_P346delinsES, and N348_I360delinsK.


In various embodiments, the D2A variant evaluated in accordance with the disclosure comprises the D2A wildtype amino acid sequence or a variant sequence described herein, fused to the C-terminus of a carrier protein, with a linking amino acid sequence. In some embodiments, the carrier protein is Fc fragment or albumin. In some embodiments, the carrier protein is human albumin. The linking sequence can be a flexible linker predominately composed of Gly, or Gy and Ser. In some embodiments, the linker is composed of Gly and amino acids having hydrophilic side chains (e.g., Ser, Thr). In some embodiments, the linker is from 5 to 20 amino acids. An exemplary linker has the structure (GGGGS)3.


The peptide linker may be a flexible linker, a rigid linker, or in some embodiments a physiologically-cleavable linker (e.g., a protease-cleavable linker). In some embodiments, the linker is 5 to 100 amino acids in length, or is 5 to 50 amino acids in length.


Linkers, where present, can be selected from flexible, rigid, and cleavable peptide linkers. Flexible linkers are predominately or entirely composed of small, non-polar or polar residues such as Gly, Ser and Thr. An exemplary flexible linker comprises (GlyySer)n linkers, where y is from 1 to 10 (e.g., from 1 to 5), and n is from 1 to about 10, and in some embodiments, is from 3 to about 6. In exemplary embodiments, y is from 2 to 4, and n is from 3 to 8. Due to their flexibility, these linkers are unstructured. More rigid linkers include polyproline or poly Pro-Ala motifs and α-helical linkers. An exemplary α-helical linker is A(EAAAK)nA, where n is as defined above (e.g., from 1 to 10, or 2 to 6). Generally, linkers can be predominately composed of amino acids selected from Gly, Ser, Thr, Ala, and Pro. Exemplary linker sequences contain at least 10 amino acids, and may be in the range of 15 to 35 amino acids. Exemplary linker designs are provided as SEQ ID NOS: 41 to 51.


In some embodiments, the variant comprises a linker, wherein the amino acid sequence of the linker is predominately glycine and serine residues, or consists essentially of glycine and serine residues. In some embodiments, the ratio of Ser and Gly in the linker is, respectively, from about 1:1 to about 1:10, from about 1:2 to about 1:6, or about 1:4. Exemplary linker sequences comprise S(GGS)4GSS (SEQ ID NO: 46), S(GGS)9GS (SEQ ID NO: 47), (GGS)9GS (SEQ ID NO: 48). In some embodiments, the linker has at least 10 amino acids, or at least 15 amino acids, or at least 20 amino acids, or at least 25 amino acids. For example, the linker may have a length of from 15 to 30 amino acids. In various embodiments, longer linkers of at least 15 amino acids can provide improvements in yield upon expression in Pichia pastoris.


In other embodiments, the linker is a physiologically-cleavable linker, such as a protease-cleavable linker. For example, the protease may be a coagulation pathway protease, such as activated Factor XII. In certain embodiments, the linker comprises the amino acid sequence of Factor XI (SEQ ID NO: 49) and/or prekallikrein (SEQ ID NO: 50 or 51) or a physiologically cleavable fragment thereof. In other embodiments, the linker includes a peptide sequence that is targeted for cleavage by a neutrophil specific protease, such as neutrophil elastase, cathepsin G, and proteinase 3.


In some embodiments, the DNASE2B (D2B) variant evaluated and selected for therapy in accordance with embodiments of this disclosure comprise an amino acid sequence that is at least 80% identical to the enzyme defined by SEQ ID NO: 7, with one or more building block substitutions.


In some embodiments, the building block substitutions to D2B are selected from non-human D2B proteins and result in variants of human D2B, which feature one or more of the following mutations: A28P, A28T, T29E, T29V, T29K, S31A, R33I, N34S, E36Y, E36D, A37P, T44I, T44A, T44V, K50R, R51Q, R51K, Q52T, N53S, N53D, N53E, K54R, E55A, E55G, S56G, G57E, G57T, G57R, T59A, T59M, E62Q, E62D, E62G, T70R, T70M, T70I, R71Q, S72T, R74N, R74S, R74K, K75R, E77L, E77H, E77K, Q78Y, Q78H, Q78L, M80I, M80V, D82T, D82S, D82A, T83S, K84R, K84D, V86A, V86S, Q92E, Q93H, E96D, A97T, Y98H, Y98N, Y98C, A99D, A99H, S100A, SF, K101E, S102T, S102N, S102D,N104D,N104S, L108V, I109L, G113A, V114I, K116G, K116A, P117S, V118A, N119T, N119G, N119S, Y120C, R122G, K123Q, K123N, Y124F, T127A, L132V, V136T, V136I, I145V, Q147K, Q147R, I151V, I151T, E154H, E154K, D157E, P160T, P160S, T161S, R164Q, N165Y, N165H, G166A, S168T, S168A, S168N, I170L, I170M, F174L, K175G, K175R, N178S, Y179F, A181E, A181T, S184F, V188I, C189L, C189F, C189Y, N190Q, V192I, S195R, S197F, A200S, A200N, A200T, T201I, T201A, H203R, Q204W, Q204M, E205K, 1207V, 1207F, H208Y, H208Y, M209L, Q211R, L212M, T214A, R215K, R215G, A216S, S217T, S217H, S218A, S219L, E220K, G223V, G223S, R224Q, L225Y, L225R, L225H, T227A, T228E, T228V, T228S, Q230H, Q233R, Q235L, K236N, K236S, L238V, L238I, S243F, D244S, D244T, S245F, F246Y, L247T, L247H, A252T, A252V, A253G, M255I, R258K, R258H, R258Q, T261V, T265A, T265V, E266Q, T267S, R270K, R272K, R272N, R272G, Q273H, Y283H, C285I, I288V, A290S, K292G, K292R, L293V, L293G, L293I, R295G, R295S, R295L, R295H, H296K, H296Q, Y298D, S300P, Y302R, Y302H, Q303H, A306S, I310V, Q312I, Q312T, Q312R, Q312L, G314D, G314R, T315S, K316A, K316Q, N317A, R318H, P329L, H330Y, F333L, F333S, S335G, T341S, T341N, Q342K, W344H, W344R, W344Q, Q345H, Q345Y, Q345R, Q345N, Q349H, Q351H, Q351D, Q351E, G352K, G352R, V354Y, L355S, Y356R, Y356H, Y357H, E358G, E358A, S359F, S359N, S359D, and K361N.


In others embodiments, the building block substitutions to D2B are selected from human DNASE2A (D2A) and result in variants of human D2B, which feature one or more of the following mutations: K2_M6delinsIPLLL, R8A, R11_G21delinsCVP, A28_S31delinsGALT, R33_E36delinsYGDS, K38_A39delinsQP, T44_F45delinsVV, K50delinsAL, Q52_K54delinsGSG, S56_T59delinsAAQR, E62Q, L64K, S70_S72delinsESSGG, K75_Q78delinsDGRA, M80I, D82_K87delinsSPEGAV, T90S, Q93P, E96_Y99delinsR, K101_T105delinsNTSQL, Y107F, I109L, G113_V114delinsQP, K106Q, V118_Y120delinsSKAQD, L130V, W133_Q137delinsLDHDG, I142V, I145V, Q147N, F148_P149insP, I15I_G155delinsASSAA, D157 Y158delinsSW, P160_N165delinsHSACTY, S168_I170delinsTLL, I172_T173delinsVS, K175_N177delinsPFA, Y179_s184delinsFSKMGK, L187_N190delinsTYTY, N192W, S195_T201delinsNYQLEGI, H203A, L206_S218delinsFPDLENVVKGHHV, E220_I221delinsQE, G223_T227delinsWNSSI, A232_Q233delinsQA, Q236_K236delinsAV, L238_H239delinsQS, S243_S2456delinsFSK, L247G, 1250_A253delinsLYSG, M255L, Q257_R258delinsAA, K260G, H262N, L264_T267delinsQVQF, Q269_E274delinsHKTVGI, L281_Y284delinsDIWQ, Y286L, 1288_A290delinsVNQ, K292_Y298delinsAFPGAGPS, S300N, Y302_Q303delinsTE, A306S, 1310V, Q312P, T315_R318delinsP, I322V, L325M, S328_F333delinsNQGEEQ, S335G, F338 I339delinsTL, N343_Q348delinsLPALWK, G353P, L355_Y356delinsKN, E358_S359delinsQP, and K361 delinsNGMARKPSRAYKI.


In various embodiments, the D2B variant evaluated in accordance with the disclosure comprises the D2B wildtype amino acid sequence or a variant sequence described herein, fused to the C-terminus of a carrier protein, with a linking amino acid sequence. In some embodiments, the carrier protein is Fc fragment or albumin. In some embodiments, the carrier protein is human albumin. The linking sequence can be a flexible linker predominately composed of Gly, or Gy and Ser. In some embodiments, the linker is composed of Gly and amino acids having hydrophilic side chains (e.g., Ser, Thr). In some embodiments, the linker is from 5 to 20 amino acids. An exemplary linker has the structure (GGGGS)3.


The peptide linker may be a flexible linker, a rigid linker, or in some embodiments a physiologically-cleavable linker (e.g., a protease-cleavable linker). In some embodiments, the linker is 5 to 100 amino acids in length, or is 5 to 50 amino acids in length.


Linkers, where present, can be selected from flexible, rigid, and cleavable peptide linkers. Flexible linkers are predominately or entirely composed of small, non-polar or polar residues such as Gly, Ser and Thr. An exemplary flexible linker comprises (GlyySer)n linkers, where y is from 1 to 10 (e.g., from 1 to 5), and n is from 1 to about 10, and in some embodiments, is from 3 to about 6. In exemplary embodiments, y is from 2 to 4, and n is from 3 to 8. Due to their flexibility, these linkers are unstructured. More rigid linkers include polyproline or poly Pro-Ala motifs and α-helical linkers. An exemplary α-helical linker is A(EAAAK)nA, where n is as defined above (e.g., from 1 to 10, or 2 to 6). Generally, linkers can be predominately composed of amino acids selected from Gly, Ser, Thr, Ala, and Pro. Exemplary linker sequences contain at least 10 amino acids, and may be in the range of 15 to 35 amino acids. Exemplary linker designs are provided as SEQ ID NOS: 41 to 51.


In some embodiments, the variant comprises a linker, wherein the amino acid sequence of the linker is predominately glycine and serine residues, or consists essentially of glycine and serine residues. In some embodiments, the ratio of Ser and Gly in the linker is, respectively, from about 1:1 to about 1:10, from about 1:2 to about 1:6, or about 1:4. Exemplary linker sequences comprise S(GGS)4GSS (SEQ ID NO: 46), S(GGS)9GS (SEQ ID NO: 47), (GGS)9GS (SEQ ID NO: 48). In some embodiments, the linker has at least 10 amino acids, or at least 15 amino acids, or at least 20 amino acids, or at least 25 amino acids. For example, the linker may have a length of from 15 to 30 amino acids. In various embodiments, longer linkers of at least 15 amino acids can provide improvements in yield upon expression in Pichia pastoris.


In other embodiments, the linker is a physiologically-cleavable linker, such as a protease-cleavable linker. For example, the protease may be a coagulation pathway protease, such as activated Factor XII. In certain embodiments, the linker comprises the amino acid sequence of Factor XI (SEQ ID NO: 49) and/or prekallikrein (SEQ ID NO: 50 or 51) or a physiologically cleavable fragment thereof. In other embodiments, the linker includes a peptide sequence that is targeted for cleavage by a neutrophil specific protease, such as neutrophil elastase, cathepsin G, and proteinase 3.


In some embodiments, the DNase variant (e.g., a variant of DNASE1 (D1), DNASE1-LIKE 1 (D1L1), DNASE1-LIKE 2 (D1L2), DNASE1-LIKE 3 Isoform 1 (D1L3), DNASE1-LIKE 3 Isoform 2 (D1L3-2), DNASE2A (D2A), and DNASE2B (D2B)) comprises an N-terminal or C-terminal fusion to a half-life extending moiety, such as albumin, transferrin, an Fc, or elastin-like protein. See U.S. Pat. No. 9,458,218, which is hereby incorporated by reference in its entirety. In some embodiments, the DNase variant is dimerized by an immunoglobulin hinge region. For example, the engineered enzymes described herein may also include an Fc-fusion domain (e.g. a hinge and CH2 domains and CH3 domains of an immunoglobulin). In other cases, the engineered DNase variant is fused to albumin, e.g., human albumin (SEQ ID NO: 12) or a fragment thereof. See WO 2015/066550; U.S. Pat. No. 9,221,896, which are hereby incorporated by reference in its entirety. Albumin can be fused at the N-terminus or the C-terminus of the engineered DNase variant, and may optionally comprise an amino acid linker. In some embodiments, two DNase variants are dimerized by an Fc hinge region, creating a dimeric molecule with synergistic functional properties for degrading NETs.


In some embodiments, human albumin and a flexible linker is fused to the N-terminus of DNASE1 (e.g., SEQ ID NO: 13), DNASE1-LIKE 1 (e.g., SEQ ID NO: 14), DNASE1-LIKE 2 (e.g., SEQ ID NO: 15), DNASE1-LIKE 3 Isoform 1 (e.g., SEQ ID NO: 16), DNASE1-LIKE 3 Isoform 2 (e.g., SEQ ID NO: 17), DNASE2A (e.g., SEQ ID NO: 18), and DNASE2B (e.g., SEQ ID NO: 19).


In some embodiments, the recombinant DNase variant comprises one or more polyethylene glycol (PEG) moieties, which may be conjugated at one or more of positions or the C-terminus. In some embodiments, the native amino acid at that position is substituted with an amino acid having a side chain suitable for crosslinking with hydrophilic moieties, to facilitate linkage of the hydrophilic moiety to the peptide. In other embodiments, an amino acid modified to comprise a hydrophilic group is added to the peptide at the C-terminus. The PEG chain(s) may have a molecular weight in the range of about 500 to about 40,000 Daltons. In some embodiments, the PEG chain(s) have a molecular weight in the range of about 500 to about 5,000 Daltons. In some embodiments, the PEG chain(s) have a molecular weight of about 10,000 to about 20,000 Daltons.


The extracellular DNase variants can be screened in assays for altered properties, including altered pH and temperature optimum, requirement for divalent cations for enzymatic activity, mechanisms of enzymatic inhibition (e.g. salt, divalent cations, actin, heparin, proteases), substrate affinity and specificity (e.g. single-stranded DNA, double-stranded DNA, chromatin, NETs, plasmid DNA, mitochondrial DNA), localization upon secretion (e.g. membrane-bound, extracellular matrix), localization signals (e.g. nuclear localization signal, membrane anchor), glycosylation sites, disulfide-bonds and unpaired cysteines, compatibility with GMP-compliant in vitro expression systems (e.g. bacteria. yeast, mammalian cells), compatibility with carriers (e.g. PEGylation, Fc fragment, albumin), compatibility with GMP-compliant purification methods (e.g. anion exchange resins, cation exchange resins), toxicological profile, tissue penetration, pharmacokinetics and pharmacodynamics.


In some embodiments, the DNase variants are evaluated using an in vitro nucleic acid degradation assay, which can employ single or double-stranded DNA, plasmid DNA, mitochondrial DNA, NETs, or may employ chromatin. In some embodiments, the assay is a NET-degrading assay. The in vitro assay can be performed under different conditions including varying pH, temperature, divalent cations, and/or salt, to evaluate the enzyme characteristics for clinical applications. In some embodiments, enzyme activity is evaluated with fusion to carrier proteins such as albumin or Fc, or with PEGylation.


In some embodiments, the DNase variants are evaluated for their expression potential in prokaryotic and/or eukaryotic (including mammalian and non-mammalian) expression systems, including their ease of expression, yield of recombinant enzyme, ability to be secreted as active protein, the lack of inclusion bodies, the presence of and identification of sites of glycosylation, and ease of purification with or without purification tags. In some embodiments, enzyme expression is evaluated with fusion to carrier proteins such as albumin or Fc. In some embodiments, DNase variants are evaluated with substitution of any unpaired Cysteines.


In some embodiments, the DNase variants are evaluated for short term and/or long term stability (e.g., upon storage for several months at 4° C. and/or room temperature). Stability can be evaluated by formation of aggregates, change of composition color, and/or enzyme activity.


In some embodiments, the DNase variants are evaluated in animal models, including for immunogenic potential (e.g., presence of anti-DNase variant antibodies), half-life in circulation, protease resistance, bioavailability, and/or NET-degrading activity. In some embodiments, activity is evaluated in disease models. Exemplary animal models may include rodent models (mouse, rat, rabbit) or primate models (e.g., chimpanzee).


In some embodiments, at least one DNase variant is evaluated in a genetically modified mouse deficient in D1 and D1L3 activity, the mouse further having a heterologous expression of a G-CSF polynucleotide (e.g., in hepatocyte cells) or induction of a sustained endogenous G-CSF expression (e.g., via repetitive administration of microbial compounds). This mouse model accumulates NETs and rapidly develops NET-related vascular occlusions. In these embodiments, the invention comprises selecting DNase enzyme that reduces the accumulation of NETs. The selected enzyme is formulated (as described) for administration to a human patient. One skilled in the art recognizes standard methods for generating double knockout Dnase1−/−, Dnase113−/−mice. Detailed descriptions can be found in, for example, U.S. Application Publication No. US 2019/0350178 and PCT International Patent Publication No. WO 2019/036719, the disclosure of which is incorporated herein by reference the in its entirety.


The invention further provides pharmaceutical compositions comprising extracellular DNase variant as described herein, or optionally the polynucleotide or the vector as described, and a pharmaceutically acceptable carrier. The pharmaceutical composition may be formulated for any administration route, including topical, parenteral, or pulmonary administration. In various embodiments, the composition is formulated for intradermal, intramuscular, intraperitoneal, intraarticular, intravenous, subcutaneous, intraarterial, oral, sublingual, pulmonary, or transdermal administration.


In various embodiments, a selected DNase variant is formulated with a “pharmaceutically acceptable carrier”, which includes any carrier that does not interfere with the effectiveness of the biological activity and is not toxic to the patient to whom it is administered. Examples of suitable pharmaceutical carriers are well known in the art and include phosphate buffered saline solutions, water, emulsions, such as oil/water emulsions, various types of wetting agents, sterile solutions etc. Such carriers can be formulated by conventional methods and can be administered to the subject at a suitable dose. Preferably, the compositions are sterile. These compositions may also contain adjuvants such as preservative, emulsifying agents and dispersing agents. Prevention of the action of microorganisms may be ensured by the inclusion of various antibacterial and antifungal agents.


In other aspects, the invention provides a method for treating a subject in need of extracellular DNA degradation, extracellular chromatin degradation, extracellular trap (ET) degradation and/or neutrophil extracellular trap (NET) degradation. The method comprises administering a therapeutically effective amount of the extracellular DNase variant or composition described herein. Exemplary indications where a subject is in need of extracellular DNA or chromatin degradation (including ET or NET degradation) are disclosed in PCT/US18/47084 (corresponding to WO 2019/036719 and U.S. Pat. No. 10,696,956).


In some embodiments, the subject exhibits impaired NET degradation and/or exhibits pathological NET accumulation. In some embodiments, the subject has a chronic or acute inflammatory disorder. In some embodiments, the subject has an acute or chronic infection.


In various embodiments, the present invention pertains to the treatment of diseases or conditions characterized by the presence or accumulation of NETs. Such diseases or conditions include, but are not limited to, diseases associated with chronic neutrophilia (e.g., an increase in the number of neutrophils), neutrophil aggregation and leukostasis, thrombosis and vascular occlusion (e.g. sickle cell disease), ischemia-reperfusion injury (e.g. midgut volvulus, testicular torsion, limb ischemia reperfusion, vital organ ischemia-reperfusion, organ transplantation), surgical and traumatic tissue injury, an acute or chronic inflammatory reaction or disease, an autoimmune disease (e.g. systemic lupus erythematosus (SLE), lupus nephritis, rheumatoid arthritis, vasculitis, systemic sclerosis), cardiovascular disease (e.g., myocardial infarction, stroke, atherosclerosis, venous thromboembolism, including thrombolytic therapy), metabolic disease (e.g., diabetes), systemic inflammation (e.g., systemic inflammatory response syndrome (SIRS), sepsis, septic shock, disseminated intravascular coagulation (DIC), and thrombotic microangiopathy (TMA)), inflammatory diseases of the respiratory tract (e.g. cystic fibrosis, chronic obstructive pulmonary disease (COPD), acute lung injury (ALI), smoke induced lung injury, transfusion induced lung injury (TRALI), acute respiratory distress syndrome (ARDS), and asthma, atelectasis, bronchitis, empyema), renal inflammatory diseases (acute and chronic kidney diseases, including acute kidney injury (AKI) and chronic kidney disease (CKD), inflammatory diseases related to transplated tissue (e.g. graft-versus-host disease) and cancer (e.g. leukemia, tumor metastasis, and solid tumors).


In some embodiments, the subject has or is at risk of NETs occluding ductural systems. The present invention can be administered to a subject to treat pancreatitis, cholangitis, conjunctivitis, mastitis, dry eye disease, obstructions of vas deferens, or renal diseases. In some embodiments, the ductal system is bile duct, tear duct, lactiferous duct, cystic duct, hepatic duct, ejaculatory duct, parotid duct, submandicular duct, major sublingual duct, bartholin's duct, cerebral aqueduct, pancreas, mammary gland, vas deferens, ureter, urinary bladder, gallbladder, and liver. For example, the subject may have pancreatitis, cholangitis (e.g., primary sclerosing cholangitis), conjunctivitis, mastitis, dry eye disease, an obstruction of the vas deferens, or renal disease. In some embodiments, the DNase enzyme is administered by intravenous, intraarterial, or intraperitoneal administration. In various embodiments, the DNase when applied, for example, intravenously, will be present in enzymatically active form in various ductal systems, such as in bile fluid.


In some embodiments, the subject has or is at risk of NETs accumulating on endothelial surfaces (e.g., surgical adhesions), the skin (e.g. wounds/scarring), or in synovial joints (e.g. gout and arthritis). The present invention can be administered to a subject to treat a condition characterized by an accumulation of NETs on an endothelial surface such as, but not limited to, a surgical adhesion. In various embodiments, the present invention can be administered to a subject to treat a condition characterized by an accumulation of NETs on skin such as, but not limited to, wounds and scars. In certain embodiments, the present invention can be administered to a subject to treat a condition characterized by an accumulation of NETs in a synovial joint such as, but not limited to, gout and arthritis.


In various embodiments, the subject has a disease that is or has been treated with wild-type DNases, including Dl and streptodornase. Such diseases or conditions include thrombosis, stroke, sepsis, lung injury, atherosclerosis, viral infection, sickle cell disease, myocardial infarction, ear infection, wound healing, liver injury, endocarditis, liver infection, pancreatitis, primary graft dysfunction, limb ischemia reperfusion, kidney injury, blood clotting, alum-induced inflammation, hepatorenal injury, pleural exudations, hemotorax, intrabiliary blood clots, post pneumatic anemia, ulcers, otolaryngological conditions, oral infections, minor injuries, sinusitis, post-operative rhinoplasties, infertility, bladder catheter, wound cleaning, skin reaction test, pneumococcal meningitis, gout, leg ulcers, cystic fibrosis, Kartegener's syndrome, asthma, lobar atelectasis, chronic bronchitis, bronchiectasis, lupus, primary cilliary dyskinesia, bronchiolitis, empyema, pleural infections, cancer, dry eyes disease, lower respiratory tract infections, chronic hematomas, Alzheimer's disease, and obstructive pulmonary disease.


In certain embodiments, the present invention pertains to the treatment of diseases or conditions characterized by deficiency of D1, deficiency of D1L3, and deficiency of D1 and D1L3. In some cases, the subject has a mutation in the Dnase1 and/or the Dnasell3 gene. Such subjects can also have an autoimmune disease (e.g., SLE, systemic sclerosis) or an inflammatory disease. In some cases, the subject has an acquired inhibitor of Dl (e.g., anti-DNase1 -antibody and actin) and/or the D1L3 (e.g., anti-DNasell3-antibody). Such subjects can also have an autoimmune disease (e.g., SLE, systemic sclerosis) or an inflammatory disease (e.g., sepsis and ischemia-reperfusion injury).


The invention is further described with reference to the following non-limiting examples.


EXAMPLES
Example 1
The Approach Used for Engineering DNase Variants for Therapeutic Applications

DNASE1 (D1) forms along with DNASE1-LIKE 1 (D1L1), DNASE1-LIKE 2 (D1L2) and DNASE1-LIKE 3 (D1L3), the DNASE1-protein family, a group of homologous secreted DNase enzymes. DNASE2A and DNASE2B form an additional group of homologous DNase enzymes. DNASE1- and DNASE2-protein family members are evolutionary conserved and expressed in various species, including humans. In general, all extracellular DNase enzymes provide drug candidates for therapies of diseases that are associated NETs. However, the physical, enzymatic, toxicological, and pharmacokinetic properties of these enzymes are not ideal for clinical applications.


An engineered D1 variant that is resistant to actin has been generated. Actin is an inhibitor of wild type D1. In brief, the 3D structure of the actin-DNASE1 complex was generated and actin binding sites in D1 were identified. Next, recombinant D1 variants with amino acids substitutions in the actin binding sites were expressed and tested for their sensitivity towards actin inhibition. The mutation A136F in SEQ ID NO: 1 was identified to generate the best actin-resistant D1 variants. See Ulmer et al., PNAS USA Vol. 93, pp 8225-8229 (1996).


Rats express a D1 variant that is naturally resistant to actin inhibition due to mutations in actin binding sites. Furthermore, the enzymatic activity of human D1L2 and D1L3 is not inhibited by actin. Indeed, human D1L3 features an F139, which corresponds to A136 in human D1 and likely causes the actin-resistance of D1L3.


Without being bound by theory, it was proposed that enzymatic properties that are favorable for development of therapy with extracellular DNase enzymes can be transferred to human extracellular DNase enzymes from extracellular DNase enzymes expressed in other species (e.g. rat) or from other members of the same extracellular DNase protein family (e.g. DNASE1-protein family comprised of DNASE1 (D1), DNASE1-LIKE 1 (D1L1), DNASE1-LIKE 2 (D1L2), DNASE1-LIKE 3 Isoform 1 (D1L3), DNASE1-LIKE 3 Isoform 2 (D1L3-2), and DNASE1-protein family comprised of DNASE2A (D2A), and DNASE2B (D2B)).


A protein engineering technology, termed Building Block Protein Engineering, can be applied to members of the DNASE1 and DNASE2 protein family and an extracellular DNase (e.g. D1, D1L1, D1L2, D1L3, D1L3-2, D2A, D2B). Building Block Protein Engineering is based on the following steps: providing a protein-protein alignment of donor and recipient DNase enzymes; identifying variable amino acid(s) for transfer, the variable amino acid(s) being flanked by one or more conserved amino acids in the donor and recipient DNase enzymes; substituting the variable amino acid(s) of the recipient DNase with the variable amino acid(s) of the donor DNase to create a chimeric DNase; and recombinantly producing the chimeric DNase.


This approach can generate two distinct types of libraries with variants of extracellular DNase enzymes: a library based on phylogenetic variation of a human extracellular DNase, and a library that is based on variation among DNase-family members (FIG. 1).


For example, FIG. 2 shows the alignment of human D1L3, with D1L3 from other species, including chimpanzee, baboon, mouse, rat, rabbit, dog, pig, guinea pig, cow, and elephant, and which identifies non-conserved amino acids (Building Blocks) that when transferred to human D1L3 result in phylogenetic variants of human D1L3. These feature the following mutations: M21L, K22R, I22L, I22V, E33A, E33Y, E33G, S34A, S34T, Q36K, Q36R, E37A, E37Q, D48N, K39Q, K39H, K39R, K39C, N40E, N40Q, N40K, A41V, V44I, V53I, I53L, I54M, V57L, I60V, N64S, H64S, R66N, R66M, I70V, I70M, I70T, M72L, E73K, K74R, R77G, R81K, G82S, I83V, I83T, T84M, T84K, S91P, T91V, T91A, L105V, K107M, V111L, S112T, R115T, R115A, R115K, R115D, R115Q, S116K, S116N, S116Y, H118L, H118V, Y119F, H120G, Y122N, Q123E, D124A, D124S, D124N, G125E, A127V, A127T, V129A, F135Y, V137T, Q140H, S141A, H143F, H143Y, V146A, I152V, T157S, T160A, V162I, K163R, V169A, E170D, T173M, T173L, V175M, K176R, K176Q, H177S, H177R, R178Q, K180E, K180N, K181T, K181V, E183A, E183Q, A201S, K203Q, K203R, R212K, R212N, R212G, R212M, V214I, G218K, G218A, Q220E, Q220D, K227R, K227S, K227E, N239K, N239S, N239H, R239C, Q241P, E242D, E242N, V244I, S245N, S245R, K250R, K250D, K250R, K250G, K250N, K250Q, N252S, S253G, S253L, V254T, V254I, D256N, Q258R, Y261F, K262D, K262E, K262L, K262R, K262Q, T264S, E266S, E267K, E267Q, E267K, D270N, D270E, V271I, S282E, R285T, F287I, S290N, K291R, V294I, T295S, T295Q, L296V, L296P, L296S, R297K, K299R, T300K, T300A, S302G, S302A, S302V, S302T, K303N, K303S, K303R, R304H, R304S, S305P, S305T, and S305A.



FIG. 3 shows the alignment of human D1L3 with human D1L1, and identifies non-conserved amino acids (Building Blocks) that when transferred to human D1L3 result in variants of human D1L3. These feature the following mutations: S2_L5delinsHYPT, P7_L9delinsL, L11F, L13 S17delinsILANG, L19delinsQ, M21F, S25A, V28_S34delinsAQRLTLA, Q36_A41delinsVAREQV, V44_I45delinsTL, K47_K50delinsRILA, 155_M58delinsMVLQ, I60_K61delinsVV, N6_C68delinsSGSAI, 170L, M72_L74delinsLRE, N78_R81delinsFDGS, 183_T84delinsP, N86_I89delinsSTLS, S91_R92delinsPQ, N96S, K99M, Q101T, A103_L105delinsVYF, K107_S112delinsRSHKTQ, K114_R115delinsLS, H118V, H120N, Y122_A127delinsED, S131A, V137_W138delinsAQ, Q140_S141delinsSL, H143_F149delinsSNVLPSL, I151_I152delinsLV, E159_V162delinsKAVE, I165_A167delinsLNA, V169_E170delinsYD, Y172_D174delinsFLE, K176_R178delinsSQH, K180_F184delinsQSKDV, G193D, S195_P198delinsASLT, A201_R206delinsRLDKLE, D210E, R212G, V214H, L216V, G218A, Q220G, K226_K227delinsRA, N230H, A232T, I236V, R239H, Q246_V249delinsERCR, S246_V254delinsLLHTAAA, Q258_K262delinsPTSFQ, D270_V271delinsNI, F275Y, F279_K280delinsVE, Q282_S283delinsKL, R285Q, F287_K292delinsHSVQPL, V294L, L296_S205delinsVLLLLSLLSPQLCPAA.


Such extracellular DNase variants can be screened in assays for altered properties, including altered pH and temperature optimum, requirement for divalent cations for enzymatic activity, mechanisms of enzymatic inhibition (e.g. salt, divalent cations, actin, heparin, proteases), substrate affinity and specificity (e.g. single-stranded DNA, double-stranded DNA, chromatin, NETs, plasmid DNA, mitochondrial DNA), localization upon secretion (e.g. membrane-bound, extracellular matrix), localization signals (e.g. nuclear localization signal, membrane anchor), glycosylation sites, disulfide-bonds and unpaired cysteines, compatibility with GMP-compliant in vitro expression systems (e.g. bacteria. yeast, mammalian cells), compatibility with carriers (e.g. PEGylation, Fc fragment, albumin), compatibility with GMP-compliant purification methods (e.g. anion exchange resins, cation exchange resins), toxicological profile, tissue penetration, pharmacokinetics and pharmacodynamics.


An extracellular DNase with altered profile can provide a drug candidate for diseases that are associated with NETs.


Example 2
Development of Building Block Engineering of DNasel-Protein Family Members

D1L3 features three sites that contain additional amino acids: the C-terminal tail starting after Q282 (NH2-SSRAFTBSKKSVTLRKKTKSKRS-COOH) (SEQ ID NO: 21), and at two sites within the enzyme at S79/R80 and at K226. The 23 amino acids of the C-terminal tail of D1L3 have been attached to the C-terminus of D1. It was observed that the insertion of an arginine-residue at position 226 of DNase1 (A226_T227insK) generated a D1-variant with reduced enzymatic activity to degrade dsDNA, while no such effect was observed with the substitution T227K. Thus, an insertion of a K/R-residue goes along with a risk of reducing D1 function. The insertion of a charged amino acid may influence the local protein structure. Given that D1 is a globular enzyme that comprises one amino acid chain, it is conceivable that such local alteration may render the whole enzyme inactive. Indeed, numerous non-conservative mutations throughout the D1 amino acid sequence inactivate the enzyme. Without being bound by theory, it was hypothesized that the transfer of local protein structures by implanting not only single arginine and lysine residues but also the neighboring amino acids sequences reduces the risk of inactivation. Conserved amino acids were searched within D1 and D1L3 for in the vicinity of A226 T227insK that can be used as anchors for the insertion. A D223/T224/T225 motif and a conserved T229 in D1 as N-terminal and C-terminal anchors, respectively were identified. 3 amino acids within D1 (ATP) were replaced with 4 amino acids, including K226, from D1L3 (VKKS) in silico. Expression of the cDNA of the new D1-variant (A226_P228delinsVKKS) in HEK239 cells revealed a functionally active enzyme with a similar dsDNA-degrading activity, when compared to wild-type D1. The data suggest that the variable amino acids between conserved amino acids are interchangeable between D1 and D1L3.


We conceptualized a building block-technology to transfer enzymatic properties from one member of the Dl-protein family to another. The following cardinal steps characterize the technology (FIG. 4):

    • (1) Provide protein-protein alignment of donor and recipient DNase
    • (2) Identify variable amino acid or amino acid sequence for transfer (building block)
    • (3) Identify conserved amino acids in donor and recipient DNase that are located up and downstream of building block (anchors), respectively.
    • (4) Replace cDNA encoding for building block between C- and N-anchors in recipient DNase, with cDNA between the anchors in donor DNase.
    • (5) Synthesize cDNA of chimeric DNase, followed by in vitro/in vivo expression into a recipient organism that is preferably deficient in both donor and recipient DNase (e.g. CHO cells or Dnase1−/−Dnase1−/−mice).


Example 3
Engineering DNase1 Variants Through Building Block Technology

A multiple-species alignment of D1 and D1L3 from human, mouse, rat, and chimpanzee (FIG. 5), showed that N- and C-terminal anchors are conserved among these species. These anchor amino acids or amino acid sequences flank 62 building blocks of variable amino acids and amino acid sequences, which include the amino acid sequence in D1 (ATP) and D1L3 (VKKS) from building blocks #49 (FIGS. 6A-6B).


The transfer of these building blocks from D1L3 into D1 generates D1-variants with the following mutations (FIGS. 6A-6B):

  • 1M_S22delinsMSRELAPLLLLLLSIHSALA, L23_A27delinsMRICS,
  • I30_T32delinsVRS, E35_T36delinsES, M38_I47delinsQEDKNAMDVI,
  • Q49_S52delinsKVIK, Y54C, 156_Q60delinsIILVM, V62_R63delinsIK,
  • S65_K72delinsSNNRICPI, L74_N76delinsMEK, Q79_T84delinsRNSRRGIT, H86N,
  • V88_V89delinsVI, E91_P92delinsSR, N96_S97delinsNT, R101Q, L103A, V105L,
  • R107_Q110delinsKEKL, A113_S116delinsVKRS, Y118H, D120H,
  • G122_N128delinsYQDGDA, T130S, N132S, A136_I137delinsFV, R139W,
  • F141_F144delinsQSPH, E146_E149delinsAVKD, A151V, V153I,
  • A157_A158delinsTT, G160_A162delinsETS, A164K, A168E, Y170 D171delinsVE,
  • L174T, Q177_K179delinsKHR, G181L182delinsKA, D184_L187delinsNFIF,
  • R199_Q202delinsPKKA, S204_S205delinsKN, W209R, S211D, T213R, Q215V,
  • P219G, S221_A222delinsQE, A226_P228delinsVKKS, H230N, V238_A239delinsLR,
  • M241_A246delinsQEIVSS, D250K, A252_P254delinsNSV, N256D,
  • A259_A260delinsKA, G262K, S264_L267delinsTEEE, Q269_I271delinsLDV,
  • Y275F, V279_M280delinsFK, and K282delinsQSSRAFTNSKKSVTLRKKTKSKRS.


The following D1L3-variants are generated if the building blocks are transferred from D1 to D1L3 (FIGS. 6A-6B):

  • M1_A20delinsMRGMKLLGALLALAALLQGAVS, M21_S25delinsLKIAA,
  • V28_S30delinsIQT, E33_S34delinsET, Q36_I45delinsMSNATLVSYI,
  • K47_K50delinsQILS, C52Y, I54_M58delinsIALVQ, 160_K61delinsVR,
  • S63_I70delinsSHLTAVGK, M72_K74delinsLDN, R77_T84delinsQDAPDT, N86H,
  • V88_I89delinsVV, S91_R92delinsEP, N96_T97delinsNS, Q101R, A103L, L105V,
  • K107_L110delinsRPDQ, V113_S116delinsAVDS, H118Y, H120D,
  • Y122_A127delinsGCEPCGN, V129T, S131N, 135F_136VdelinsAI, W138R,
  • Q140_H143delinsFSRF, A145_D148delinsAVKD, V150A, I152A,
  • T156_T157delinsAA, E159_S161delinsGDA, K163A, E167A, V169_E170delinsYD,
  • T173L, K176_R178delinsQEK, K180_A181delinsGL, N183_F186delinsDVML,
  • P198_A20delinsRPSQ, K203_N204delinsSS, R208W, D210S, R212T, V214Q,
  • G218P, Q220_E221delinsSA, V225_S228delinsATP, N230H, L238_R239delinsVA,
  • Q241_S246delinsMLLRGA, K250D, N252_V254delinsALP, D256N,
  • K259_A260delinsAA, K262G, T264_E267delinsSDQL, L269_V271delinsVA,
  • F275Y, F279_K280delinsVM, and Q282_S305delinsK.


Next, we conceptualized a sequential approach to engineer D1-variants with D1L3 activity that starts with the transfer of multiple adjacent building blocks (clusters), continues with the transfer of individual building blocks, and ends with a transfer of individual amino acids or the combination of multiple building blocks into new chimeric enzymes (FIG. 7). This approach reduces the number of D1-D1L3-chimera in the initial screening.


To test our method, we designed a total of 19 D1-variants comprising either individual building blocks or clusters of building block cluster from D1L3 (FIG. 6). These D1-variants feature the following amino acid mutations:

  • 1M_S22delinsMSRELAPLLLLLLSIHSALA,
  • L23_A27delinsMRICS/I30_T32delinsVRS/E35_T36delinsES,
  • M38_I47delinsQEDKNAMDVI, Q49_S52delinsKVIK/Y54C/I56_Q60delinsIILVM,
  • V62_R63delinsIK/S65_K72delinsSNNRICPI/L74_N76delinsMEK,
  • Q79_T84delinsRNSRRGIT, H86N/V88_V89delinsVI/E91_P92delinsSR,
  • N96_S97delinsNT/R101Q/L103A/V105L,
  • R107_Q110delinsKEKL/A113_S116delinsVKRS/Y118H/D120H,
  • G122_N128delinsYQDGDA/T130S/N132S, A136_I137delinsFV,
  • R139W/F141_F144delinsQSPH/E146_E149delinsAVKD/A151VN153I/A157_A158d
  • elinsTT/G160_A162delinsETS/A164K, A168E/Y170_D171 delinsVE/L174T,
  • Q177_K179delinsKHR/G181_L182delinsKA/D184_L187delinsNFIF,
  • R199_Q202delinsPKKA/S204_S205delinsKN/W209R/S211D/T213R/Q215V/P219G/
  • S221_A222delinsQE, A226_P228delinsVKKS,
  • H230N/V238_A239delinsLR/M241_A246delinsQEIVSS/D250K/A252_P254delinsNS
  • N256D/A259_A260delinsKA/G262K/S264_L267delinsTEEE/Q269_I271delinsLDV,
  • and Y275F/V279_M280delinsFK/K282delinsQSSRAFTNSKKSVTLRKKTKSKRS.


Next, we cloned the cDNA into an expression vector, which was transfected into HEK293 cells. Analysis of the cell supernatants showed dsDNA degradation by all samples (FIG. 8). Furthermore, we observed that the transfer of building blocks (BB) 11, BB 12-14, BB 26, BB 41-48, and BB 49 from D1L3 to D1 resulted in enzymes with increased chromatin degrading activity. All of these chimeric enzymes exhibited the same or more activity to degrade dsDNA substrates than wild-type D1. The building blocks 11 and 49 from D1L3 contain R80/R81 and K227, respectively, which are not present in D1. The D1L3-BB cluster 41-48 features 5 additional arginine and lysine residues than its counterpart in D1. These additional cationic amino acids may be responsible for the hyperactivity. The D1-building blocks 12-14 and 26 contain the amino acid sequences H86 to R95 and A136 to V138 in SEQ ID NO: 1, which includes amino acid residues that are required for binding of the D1-inhibitor actin. Thus, replacement of these amino acid sequences with the respective building blocks from D1L3, which do not interact with actin, likely generates actin-resistant variants of D1. We now combined BB 11, 14, 26, 41-19 in one novel D1-variant. We observed that the combination of these gain-of-function BBs increased the chromatin degrading of the D1 variant to levels of wild-type D1L3 (FIG. 8). Thus, the BB technology provides a robust method to generate hyperactive D1 variants.


Example 4
Expression and Characterization of D1L3 with Basic Domain Deletion (BDD) in Chinese Hamster Ovarian (CHO) Cells and in Pichia pastoris

DNASE1 and DNASE1L3 preferentially cleave protein-free DNA and DNA-histone-complexes (i.e. chromatin), respectively. Previous studies suggest that a basic domain (BD) at the C-terminus of DNASE1L3, which is absent in DNASE1, is responsible for the distinct substrate specificities of both enzymes (Sisirak et al., Cell, 2016; Keyel, Developmental Biology, 2017).


To characterize the amino acids that are responsible for chromatin-degrading activity (“chromatinase” activity), wild-type D1L3 was substituted with building block substitutions from D1, as disclosed in PCT/US2018/047084. The building block substitutions to D1L3 are selected from human D1 and result in variants of human D1L3, which feature the following mutations: M21_R22delinsLK, C24_S25delinsAA, V28_S30delinsIQT, S34T, Q36_V44delinsMSNATLVSY, K47_K50delinsQILS, C52Y, I55_M58delinsIALVQE, I60K61delinsVR, N64I70delinsHLTAVGK, M72K74delinsLDN, R77_I83delinsQDAPD, N86H, I89V, S91_R92delinsEP, T97S, Q101R, A103L, L105V, K107_L110delinsRPDQ, V113_R115delinsAVD, H118Y, H120D, Y122_A127delinsGCEPCGN, V129T, S131N, F135_V136delinsAI, W138R, Q140_H143delinFSRF, A145_D148delinsEVRE, V150A, I152V, T156_T157delinsAA, E159_S161delinsGDA, K163A, E167A, V169_E170delinsYD, T173L, K176_R178delinsQEK, K180_A181delinsGL, N183_F186delinsDVML, P198A201delinsRPSQ, K203_N204delinsSS, R208W, D210S, R212T, V214Q, G218P, Q220_E221delinsSA, V225_S228delinsATP, N230H, L238_R239delinsVA, Q241_S246delinsMLLRGA, K250D, N252_V254delinsALP, D256N, K259A, K262G, T264E267delinsSDQL, L269_V271delinsQA, F275Y, F279_K280delinsVM, Q282_S205delinsK with respect to human D1L3, Isoform 1.


These 63 D1L3variants were screened for loss or gain of chromatin-degrading activity. In brief, D1L3 variants were transiently expressed in CHO cells using an in vitro expression vector. Culture supernatants were collected and tested for chromatin-degrading activity using purified nuclei as a source of chromatin. As shown in FIG. 9, the building block substitution #63 from D1 significantly improved the degradation of high-molecular weight (HMW) chromatin to small fragments, when compared to wild-type D1L3. Building block substitution #63 causes the mutation Q282_S305delinsK, which deletes the full C-terminal BD of D1L3 from amino acid position 283 to 305 and replaces glutamine (Q) at position 282 with lysine.


Example 5
The Paired Basic Amino Acid Cleaving Enzyme Furin Regulates D1L3

The BD domain of D1L3 contains an NLS and three paired basic amino acids that are potentially responsible for the inhibitory effects on enzymatic activity (FIG. 10). The NLS is located between amino acids L286 to S302 [LRKKTKS; Reference: Q13609 (Uniprot)] and therefore absent in all identified hyperactive D1L3 mutants that lack 12 amino acids or more.


Without being bound by theory, it was hypothesized that three sets of paired basic amino acids are K291/1K292, R297/K298/K299, and K303/R304 and may serve as proteolytic cleavage sites of the Paired Basic Amino Acid Cleaving Enzyme (PACE). Furin is a well-characterized PACE, which is involved in the maturation of pro-enzymes. To test the possibility of furin generating active D1L3, and to understand the possible role of furin in activation of chromatinase activity of D1L3, furin-overexpressing CHO cells were transiently transfected with wild-type and BD-deleted D1L3 (S283_S305del mutant). CHO cells without overexpression of furin were included as control. Culture supernatants were collected and tested by western blot using an antibody that targets the N-terminus of D1L3. As shown in FIG. 11, two bands of D1L3 were detected in furin-overexpressing CHO cells. The top band corresponded to D1L3 expressed by CHO cells without overexpression of furin, whereas the lower band corresponded to the BD-deleted D1L3 (FIG. 11). The data suggest that furin and/or other proteases may directly or indirectly delete the BD and thus cause the maturation of D1L3 into its enzymatically active form. This cleavage activity further suggests that C-terminal fusion to carrier proteins will require deletions or amino acid substitutions within the BD to avoid proteolytic cleavage in vivo.


Example 6
Heterologous Expression of Wild-Type D1L3

We analyzed molecular modifications of the C-terminal BD of D1L3 following the heterologous expression of wild-type D1L3.


In the first study, we expressed a wild-type D1L3 that was linked to the Fc fragment via a flexible glycine-serine linker in CHO cells (FIG. 12A). As a control, we used an Fc fusion protein of a Basic Domain Deleted-D1L3. CHO cells were transiently transfected with either DNA construct. We collected supernatants and purified the Fc fusion proteins using protein A. Analysis of purified proteins by SDS-PAGE showed bands of approximately 65-70 kDa, which reflects the molecular weight of the full-length proteins (FIG. 12B). An additional lower molecular weight band (between 29-44 kDa) was observed in proteins purified from CHO cells expressing the WT-DNASE1L3 construct. Using LC-MS and LC/MS/MS, we identified that the N- and C-terminal amino acid sequence of the upper band corresponds to mature DNASE1L3 and Fc fragment, respectively. As seen by others, clipping of the C-terminal lysine residue of the Fc fragment was observed. Surprisingly, analysis of the lower band showed that the N-terminus of the protein contained the serine from the C-terminus of DNASE1L3, i.e. Serine 305 in SEQ ID NO: 4. Collectively these data suggest that the expression of wild-type DNASE1L3 leads to the clipping of the C-terminal serine residue.


In the second study, we expressed a wild-type D1L3 and a D1L3 linked at the N-terminus to albumin via a flexible glycine-serine linker in Pichia pastoris. We collected fermentation supernatants and purified the proteins using affinity chromatography. Surprisingly, we observed no full-length D1L3 protein with either expression construct, but various degrees of C-terminal truncations as identified by LC-MS and LC/MS/MS. The expression of wild-type D1L3 produced deletions of 13, 14, and 15 amino acids. Additional proteins with deletions of 8 and 9 amino acids were detected in samples of the wild-type D1L3 albumin fusion protein. In summary, the heterologous expression of wild-type D1L3 leads to the secretion of D1L3 variants with distinct deletions in the C-terminal basic domain. Importantly, the deletions occur after, within, or before the three clusters of basic amino acids (FIG. 13). In further studies, we expressed a wild-type D1L3 linked at the C-terminus to albumin via a flexible glycine-serine linker in Pichia pastoris and observed two proteins with a molecular weight similar to D1L3 and albumin, indicating the proteolytic cleavage of the fusion protein in the linker region that contains the BD. Collectively, our data identified the 1(291/1(292, R297/K298/K299, and K303/R304 in SEQ ID NO: 4 as sites for post-translational modifications of wild-type DNASE1L3.


INCORPORATION BY REFERENCE

All patents and publications referenced herein are hereby incorporated by reference in their entireties.













Amino Acid Sequences of




Wild-Type Human DNASES




SEQ ID NO: 1




DNASE1 (NP_005212.2):




Signal Peptide, Mature Protein:





MRGMKLLGALLALAALLQGAVSLKIAAFN









IQTFGETKMSNATLVSYIVQILSRYDIAL








VQEVRDSHLTAVGKLLDNLNQDAPDTYHY








VVSEPLGRNSYKERYLFVYRPDQVSAVDS








YYYDDGCEPCGNDTFNREPAIVRFFSRFT








EVREFAIVPLHAAPGDAVAEIDALYDVYL








DVQEKWGLEDVMLMGDFNAGCSYVRPSQW








SSIRLWTSPTFQWLIPDSADTTATPTHCA








YDRIVVAGMLLRGAVVPDSALPFNFQAAY








GLSDQLAQAISDHYPVEVMLK








DNASE1-LIKE 1 (NP_006721.1):




Signal Peptide; Mature Protein:




SEQ ID NO: 2





MHYPTALLFLILANGAQAFRICAFNAQRL









TLAKVAREQVMDTLVRILARCDIMVLQEV








VDSSGSAIPLLLRELNRFDGSGPYSTLSS








PQLGRSTYMETYVYFYRSHKTQVLSSYVY








NDEDDVFAREPFVAQFSLPSNVLPSLVLV








PLHTTPKAVEKELNALYDVFLEVSQHWQS








KDVILLGDFNADCASLTKKRLDKLELRTE








PGFHWVIADGEDTTVRASTHCTYDRVVLH








GERCRSLLHTAAAFDFPTSFQLTEEEALN








ISDHYPVEVELKLSQAHSVQPLSLTVLLL








LSLLSPQLCPAA








DNASE1-LIKE 2 (NP_001365.1):




Signal Peptide, Mature Protein:




SEQ ID NO: 3





MGGPRALLAALWALEAAGTAALRIGAFNI









QSFGDSKVSDPACGSILAKILAGYDLALV








QEVRDPDLSAVSALMEQINSVSEHEYSFV








SSQPLGRDQYKEMYLFVYRKDAVSVVDTY








LYPDPEDVFSREPFVVKFSAPGTGERAPP








LPSRRALTPPPLPAAAQNLVLIPLHAAPH








QAVAEIDALYDVYLDVIDKWGTDDMLFLG








DFNADCSYVRAQDWAAIRLRSSEVFKWLI








PDSADTTVGNSDCAYDRIVACGARLRRSL








KPQSATVHDFQEEFGLDQTQALAISDHFP








VEVTLKFHR








DNASE1-LIKE 3; Isoform 1 (NP_004935.1):




Signal Peptide, Mature Protein:




SEQ ID NO: 4





MSRELAPLLLLLLSIHSALAMRICSFNVR









SFGESKQEDKNAMDVIVKVIKRCDIILVM








EIKDSNNRICPILMEKLNRNSRRGITYNY








VISSRLGRNTYKEQYAFLYKEKLVSVKRS








YHYHDYQDGDADVFSREPFVVWFQSPHTA








VKDFVIIPLHTTPETSVKEIDELVEVYTD








VKHRWKAENFIFMGDFNAGCSYVPKKAWK








NIRLRTDPRFVWLIGDQEDTTVKKSINCA








YDRIVLRGQEIVSSVVPKSNSVFDFQKAY








KLTEEEALDVSDHFPVEFKLQSSRAFTNS








KKSVTLRKKTKSKRS








DNASE1-LIKE 3, Isoform 2 (NP_001243489.1):




Signal Peptide; Mature Protein:




SEQ ID NO: 5





MSRELAPLLLLLLSIHSALAMRICSFNVR









SFGESKQEDKNAMDVIVKVIKRCDIILVM








EIKDSNNRICPILMEKLNREKLVSVKRSY








HYHDYQDGDADVFSREPFVVWFQSPHTAV








KDFVIIPLHTTPETSVKEIDELVEVYTDV








KHRWKAENFIFMGDFNAGCSYVPKKAWKN








IRLRTDPRFVWLIGDQEDTTVKKSTNCAY








DRIVLRGQEIVSSVVPKSNSVFDFQKAYK








LTEEEALDVSDHFPVEFKLQSSRAFTNSK








KSVTLRKKTKSKRS








DNASE2A (O00115):




Signal Peptide; Mature Protein:




SEQ ID NO: 6




MIPLLLAALLCVPAGALTCYGDSGQPVDW








FVVYKLPALRGSGEAAQRGLQYKYLDESS








GGWRDGRALINSPEGAVGRSLQPLYRSNT








SQLAFLLYNDQPPQPSKAQDSSMRGHTKG








VLLLDHDGGFWLVHSVPNFPPPASSAAYS








WPHSACTYGQTLLCVSFPFAQFSKMGKQL








TYTYPWVYNYQLEGIFAQEFPDLENVVKG








HHVSQEPWNSSITLTSQAGAVFQSFAKFS








KFGDDLYSGWLAAALGTNLQVQFWHKTVG








ILPSNCSDIWQVLNVNQIAFPGPAGPSFN








STEDHSKWCVSPKGPWICVGDMNRNQGEE








QRGGGILCAQLPALWKAFQPLVKNYQPCN








GMARKPSRAYKI








DNASE2B (Q8WZ79):




Signal Peptide; Mature Protein:




SEQ ID NO: 7





MKQKMMARLLRTSFALLFLGLFGVLGAAT









ISCRNEEGKAVDWFTFYKLPKRQNKESGE








TGLEYLYLDSTTRSWRKSEQLMNDTKSVL








GRTLQQLYEAYASKSNNTAYLIYNDGVPK








PVNYSRKYGHTKGLLLWNRVQGFWLIHSI








PQFPPIPEEGYDYPPTGRRNGQSGICITF








KYNQYEAIDSQLLVCNPNVYSCSIPATFH








QELIHMPQLCTRASSSEIPGRLLTTLQSA








QGQKFLHFAKSDSFLDDIFAAWMAQRLKT








HLLTETWQRKRQELPSNCSLPYHVYNIKA








IKLSRHSYFSSYQDHAKWCISQKGTKNRW








TCIGDLNRSPHQAFRSGGFICTQNWQIYQ








AFQGLVLYYESCK








Selected Amino Acid Sequences of




Human Wild-Type DNASES




C-terminal tail of human DNASE1-




LIKE 1 (NP_006721.1):




SEQ ID NO: 8




KLSQAHSVQPLSLTVLLLLSLLSPQLCPAA








Proline-rich extension of human




DNASE1-LIKE 2 (NP_001365.1):




SEQ ID NO: 9




SAPGTGERAPPLPSRRALTPPPLPAAAQN








LVLIPL








C-terminal tail of human




DNASE1-LIKE 3; Isoform 1




(NP_004935.1):




SEQ ID NO: 10




SSRAFTNSKKSVTLRKKTKSKRS








Internal sequence of human




DNASE1-LIKE 3; Absent in




Isoform 2 (NP_004935.1):




SEQ ID NO: 11




RNSRRGITYNYVISSRLGRNTYKEQYAFL








YK








Carrier Protein




Human Albumin (P02768):




Signal Peptide + Propeptide;




Mature Protein:




SEQ ID NO: 12





MKWVTFISLLFLFSSAYSRGVFRRDAHKS









EVAHRFKDLGEENFKALVLIAFAQYLQQC








PFEDHVKLVNEVTEFAKTCVADESAENCD








KSLHTLFGDKLCTVATLRETYGEMADCCA








KQEPERNECFLQHKDDNPNLPRLVRPEVD








VMCTAFHDNEETFLKKYLYEIARRHPYFY








APELLFFAKRYKAAFTECCQAADKAACLL








PKLDELRDEGKASSAKQRLKCASLQKFGE








RAFKAWAVARLSQRFPKAEFAEVSKLVTD








LTKVHTECCHGDLLECADDRADLAKYICE








NQDSISSKLKECCEKPLLEKSHCIAEVEN








DEMPADLPSLAADFVESKDVCKNYAEAKD








VFLGMFLYEYARRHPDYSVVLLLRLAKTY








ETTLEKCCAAADPHECYAKVFDEFKPLVE








EPQNLIKQNCELFEQLGEYKFQNALLVRY








TKKVPQVSTPTLVEVSRNLGKVGSKCCKH








PEAKRMPCAEDYLSVVLNQLCVLHEKTPV








SDRVTKCCTESLVNRRPCFSALEVDETYV








PKEFNAETFTFHADICTLSEKERQIKKQT








ALVELVKHKPKATKEQLKAVMDDFAAFVE








KCCKADDKETCFAEEGKKLVAASQAALGL








Amino Acid Sequences of Human




ALBUMIN-DNASE-Fusion proteins




Albumin-Linker-DNASE1;




Signal Peptide + Propeptide,




Albumin, Flexible Linker,




mature DNASE1:




SEQ ID NO: 13





MKWVTFISLLFLFSSAYSRGVFRRDAH









KSEVAHRFKDLGEENFKALVLIAFAQY








LQQCPFEDHVKLVNEVTEFAKTCVADE








SAENCDKSLHTLFGDKLCTVATLRETY








GEMADCCAKQEPERNECFLQHKDDNPN








LPRLVRPEVDVMCTAFHDNEETFLKKY








LYEIARRHPYFYAPELLFFAKRYKAAF








TECCQAADKAACLLPKLDELRDEGKAS








SAKQRLKCASLQKFGERAFKAWAVARL








SQRFPKAEFAEVSKLVTDLTKVHTECC








HGDLLECADDRADLAKYICENQDSISS








KLKECCEKPLLEKSHCIAEVENDEMPA








DLPSLAADFVESKDVCKNYAEAKDVFL








GMFLYEYARRHPDYSVVLLLRLAKTYE








TTLEKCCAAADPHECYAKVFDEFKPLV








EEPQNLIKQNCELFEQLGEYKFQNALL








VRYTKKVPQVSTPTLVEVSRNLGKVGS








KCCKHPEAKRMPCAEDYLSVVLNQLCV








LHEKTPVSDRVTKCCTESLVNRRPCFS








ALEVDETYVPKEFNAETFTFHADICTL








SEKERQIKKQTALVELVKHKPKATKEQ








LKAVMDDFAAFVEKCCKADDKETCFAE








EGKKLVAASQAALGLGGGGSGGGGSGG









GGSLKIAAFNIQTFGETKMSNATLVSY









IVQILSRYDIALVQEVRDSHLTAVGKL








LDNLNQDAPDTYHYVVSEPLGRNSYKE








RYLFVYRPDQVSAVDSYYYDDGCEPCG








NDTFNREPAIVRFFSRFTEVREFAIVP








LHAAPGDAVAEIDALYDVYLDVQEKWG








LEDVMLMGDFNAGCSYVRPSQWSSIRL








WTSPTFQWLIPDSADTTATPTHCAYDR








IVVAGMLLRGAVVPDSALPFNFQAAYG








LSDQLAQATSDHYPVEVMLK








Albumin-Linker-DNASE1-LIKE 1;




Signal Peptide + Propeptide,




Albumin, Flexible Linker,




mature DNASE1-LIKE 1:




SEQ ID NO: 14





MKWVTFISLLFLFSSAYSRGVFRRDAH









KSEVAHRFKDLGEENFKALVLIAFAQY








LQQCPFEDHVKLVNEVTEFAKTCVADE








SAENCDKSLHTLFGDKLCTVATLRETY








GEMADCCAKQEPERNECFLQHKDDNPN








LPRLVRPEVDVMCTAFHDNEETFLKKY








LYEIARRHPYFYAPELLFFAKRYKAAF








TECCQAADKAACLLPKLDELRDEGKAS








SAKQRLKCASLQKFGERAFKAWAVARL








SQRFPKAEFAEVSKLVTDLTKVHTECC








HGDLLECADDRADLAKYICENQDSISS








KLKECCEKPLLEKSHCIAEVENDEMPA








DLPSLAADFVESKDVCKNYAEAKDVFL








GMFLYEYARRHPDYSVVLLLRLAKTYE








TTLEKCCAAADPHECYAKVFDEFKPLV








EEPQNLIKQNCELFEQLGEYKFQNALL








VRYTKKVPQVSTPTLVEVSRNLGKVGS








KCCKHPEAKRMPCAEDYLSVVLNQLCV








LHEKTPVSDRVTKCCTESLVNRRPCFS








ALEVDETYVPKEFNAETFTFHADICTL








SEKERQIKKQTALVELVKHKPKATKEQ








LKAVMDDFAAFVEKCCKADDKETCFAE








EGKKLVAASQAALGLGGGGSGGGGSGG









GGSFRICAFNAQRLTLAKVAREQVMDT









LVRILARCDIMVLQEVVDSSGSAIPLL








LRELNRFDGSGPYSTLSSPQLGRSTYM








ETYVYFYRSHKTQVLSSYVYNDEDDVF








AREPFVAQFSLPSNVLPSLVLVPLHTT








PKAVEKELNALYDVFLEVSQHWQSKDV








ILLGDFNADCASLTKKRLDKLELRTEP








GFHWVIADGEDTTVRASTHCTYDRVVL








HGERCRSLLHTAAAFDFPTSFQLTEEE








ALNISDHYPVEVELKLSQAHSVQPLSL








TVLLLLSLLSPQLCPAA








Albumin-Linker-DNASE1-LIKE 2;




Signal Peptide + Propeptide,




Albumin, Flexible Linker,




mature DNASE1-LIKE 2:




SEQ ID NO: 15





MKWVTFISLLFLFSSAYSRGVFRRDAH









KSEVAHRFKDLGEENFKALVLIAFAQY








LQQCPFEDHVKLVNEVTEFAKTCVADE








SAENCDKSLHTLFGDKLCTVATLRETY








GEMADCCAKQEPERNECFLQHKDDNPN








LPRLVRPEVDVMCTAFHDNEETFLKKY








LYEIARRHPYFYAPELLFFAKRYKAAF








TECCQAADKAACLLPKLDELRDEGKAS








SAKQRLKCASLQKFGERAFKAWAVARL








SQRFPKAEFAEVSKLVTDLTKVHTECC








HGDLLECADDRADLAKYICENQDSISS








KLKECCEKPLLEKSHCIAEVENDEMPA








DLPSLAADFVESKDVCKNYAEAKDVFL








GMFLYEYARRHPDYSVVLLLRLAKTYE








TTLEKCCAAADPHECYAKVFDEFKPLV








EEPQNLIKQNCELFEQLGEYKFQNALL








VRYTKKVPQVSTPTLVEVSRNLGKVGS








KCCKHPEAKRMPCAEDYLSVVLNQLCV








LHEKTPVSDRVTKCCTESLVNRRPCFS








ALEVDETYVPKEFNAETFTFHADICTL








SEKERQIKKQTALVELVKHKPKATKEQ








LKAVMDDFAAFVEKCCKADDKETCFAE








EGKKLVAASQAALGLGGGGSGGGGSGG









GGSLRIGAFNIQSFGDSKVSDPACGSI









IAKILAGYDLALVQEVRDPDLSAVSAL








MEQINSVSEHEYSFVSSQPLGRDQYKE








MYLFVYRKDAVSVVDTYLYPDPEDVFS








REPFVVKFSAPGTGERAPPLPSRRALT








PPPLPAAAQNLVLIPLHAAPHQAVAEI








DALYDVYLDVIDKWGTDDMLFLGDFNA








DCSYVRAQDWAAIRLRSSEVFKWLIPD








SADTTVGNSDCAYDRIVACGARLRRSL








KPQSATVHDFQEEFGLDQTQALAISDH








FPVEVTLKFHR








Albumin-Linker-DNASE1-LIKE 3;




Signal Peptide + Propeptide,




Albumin, Flexible Linker,




mature DNASE1-LIKE 3:




SEQ ID NO: 16




MMKWVTFISLLFLFSSAYSRGVFRRDAH








KSEVAHRFKDLGEENFKALVLIAFAQY








LQQCPFEDHVKLVNEVTEFAKTCVADE








SAENCDKSLHTLFGDKLCTVATLRETY








GEMADCCAKQEPERNECFLQHKDDNPN








LPRLVRPEVDVMCTAFHDNEETFLKKY








LYEIARRHPYFYAPELLFFAKRYKAAF








TECCQAADKAACLLPKLDELRDEGKAS








SAKQRLKCASLQKFGERAFKAWAVARL








SQRFPKAEFAEVSKLVTDLTKVHTECC








HGDLLECADDRADLAKYICENQDSISS








KLKECCEKPLLEKSHCIAEVENDEMPA








DLPSLAADFVESKDVCKNYAEAKDVFL








GMFLYEYARRHPDYSVVLLLRLAKTYE








TTLEKCCAAADPHECYAKVFDEFKPLV








EEPQNLIKQNCELFEQLGEYKFQNALL








VRYTKKVPQVSTPTLVEVSRNLGKVGS








KCCKHPEAKRMPCAEDYLSVVLNQLCV








LHEKTPVSDRVTKCCTESLVNRRPCFS








ALEVDETYVPKEFNAETFTFHADICTL








SEKERQIKKQTALVELVKHKPKATKEQ








LKAVMDDFAAFVEKCCKADDKETCFAE








EGKKLVAASQAALGLGGGGSGGGGSGG









GGSMRICSFNVRSFGESKQEDKNAMDV









IVKVIKRCDIILVMEIKDSNNRICPIL








MEKLNRNSRRGITYNYVISSRLGRNTY








KEQYAFLYKEKLVSVKRSYHYHDYQDG








DADVFSREPFVVWFQSPHTAVKDFVII








PLHTTPETSVKEIDELVEVYTDVKHRW








KAENFIFMGDFNAGCSYVPKKAWKNIR








LRTDPRFVWLIGDQEDTTVKKSTNCAY








DRIVLRGQEIVSSVVPKSNSVFDFQKA








YKLTEEEALDVSDHFPVEFKLQSSRAF








TNSKKSVTLRKKTKSKRS








Albumin-Linker-DNASE1-LIKE 3,




Isoform 2;




Signal Peptide + Propeptide,




Albumin, Flexible Linker,




mature DNASE1-LIKE 3




Isoform 2:




SEQ ID NO: 17





MKWVTFISLLFLFSSAYSRGVFRRDAH









KSEVAHRFKDLGEENFKALVLIAFAQY








LQQCPFEDHVKLVNEVTEFAKTCVADE








SAENCDKSLHTLFGDKLCTVATLRETY








GEMADCCAKQEPERNECFLQHKDDNPN








LPRLVRPEVDVMCTAFHDNEETFLKKY








LYEIARRHPYFYAPELLFFAKRYKAAF








TECCQAADKAACLLPKLDELRDEGKAS








SAKQRLKCASLQKFGERAFKAWAVARL








SQRFPKAEFAEVSKLVTDLTKVHTECC








HGDLLECADDRADLAKYICENQDSISS








KLKECCEKPLLEKSHCIAEVENDEMPA








DLPSLAADFVESKDVCKNYAEAKDVFL








GMFLYEYARRHPDYSVVLLLRLAKTYE








TTLEKCCAAADPHECYAKVFDEFKPLV








EEPQNLIKQNCELFEQLGEYKFQNALL








VRYTKKVPQVSTPTLVEVSRNLGKVGS








KCCKHPEAKRMPCAEDYLSVVLNQLCV








LHEKTPVSDRVTKCCTESLVNRRPCFS








ALEVDETYVPKEFNAETFTFHADICTL








SEKERQIKKQTALVELVKHKPKATKEQ








LKAVMDDFAAFVEKCCKADDKETCFAE








EGKKLVAASQAALGLGGGGSGGGGSGG









GGSMRICSFNVRSFGESKQEDKNAMDV









IVKVIKRCDIILVMEIKDSNNRICPIL








MEKLNREKLVSVKRSYHYHDYQDGDAD








VFSREPFVVWFQSPHTAVKDFVIIPLH








TTPETSVKEIDELVEVYTDVKHRWKAE








NFIFMGDFNAGCSYVPKKAWKNIRLRT








DPRFVWLIGDQEDTTVKKSTNCAYDRI








VLRGQEIVSSVVPKSNSVFDFQKAYKL








TEEEALDVSDHFPVEFKLQSSRAFTNS








KKSVTLRKKTKSKRS








Albumin-Linker-DNASE2A;




Signal Peptide + Propeptide,




Albumin, Flexible Linker,




mature DNASE2A:




SEQ ID NO: 18





MKWVTFISLLFLFSSAYSRGVFRRDAH









KSEVAHRFKDLGEENFKALVLIAFAQY








LQQCPFEDHVKLVNEVTEFAKTCVADE








SAENCDKSLHTLFGDKLCTVATLRETY








GEMADCCAKQEPERNECFLQHKDDNPN








LPRLVRPEVDVMCTAFHDNEETFLKKY








LYEIARRHPYFYAPELLFFAKRYKAAF








TECCQAADKAACLLPKLDELRDEGKAS








SAKQRLKCASLQKFGERAFKAWAVARL








SQRFPKAEFAEVSKLVTDLTKVHTECC








HGDLLECADDRADLAKYICENQDSISS








KLKECCEKPLLEKSHCIAEVENDEMPA








DLPSLAADFVESKDVCKNYAEAKDVFL








GMFLYEYARRHPDYSVVLLLRLAKTYE








TTLEKCCAAADPHECYAKVFDEFKPLV








EEPQNLIKQNCELFEQLGEYKFQNALL








VRYTKKVPQVSTPTLVEVSRNLGKVGS








KCCKHPEAKRMPCAEDYLSVVLNQLCV








LHEKTPVSDRVTKCCTESLVNRRPCFS








ALEVDETYVPKEFNAETFTFHADICTL








SEKERQIKKQTALVELVKHKPKATKEQ








LKAVMDDFAAFVEKCCKADDKETCFAE








EGKKLVAASQAALGLGGGGSGGGGSGG









GGSCYGDSGQPVDWFVVYKLPALRGSG









EAAQRGLQYKYLDESSGGWRDGRALIN








SPEGAVGRSLQPLYRSNTSQLAFLLYN








DQPPQPSKAQDSSMRGHTKGVLLLDHD








GGFWLVHSVPNFPPPASSAAYSWPHSA








CTYGQTLLCVSFPFAQFSKMGKQLTYT








YPWVYNYQLEGIFAQEFPDLENVVKGH








HVSQEPWNSSITLTSQAGAVFQSFAKF








SKFGDDLYSGWLAAALGTNLQVQFWHK








TVGILPSNCSDIWQVLNVNQTAFPGPA








GPSFNSTEDHSKWCVSPKGPWTCVGDM








NRNQGEEQRGGGTLCAQLPALWKAFQP








LVKNYQPCNGMARKPSRAYKI








Albumin-Linker-DNASE2B;




Signal Peptide + Propeptide,




Albumin, Flexible Linker,




mature DNASE2B:




SEQ ID NO: 19





MKWVTFISLLFLFSSAYSRGVFRRDAH









KSEVAHRFKDLGEENFKALVLIAFAQY








LQQCPFEDHVKLVNEVTEFAKTCVADE








SAENCDKSLHTLFGDKLCTVATLRETY








GEMADCCAKQEPERNECFLQHKDDNPN








LPRLVRPEVDVMCTAFHDNEETFLKKY








LYEIARRHPYFYAPELLFFAKRYKAAF








TECCQAADKAACLLPKLDELRDEGKAS








SAKQRLKCASLQKFGERAFKAWAVARL








SQRFPKAEFAEVSKLVTDLTKVHTECC








HGDLLECADDRADLAKYICENQDSISS








KLKECCEKPLLEKSHCIAEVENDEMPA








DLPSLAADFVESKDVCKNYAEAKDVFL








GMFLYEYARRHPDYSVVLLLRLAKTYE








TTLEKCCAAADPHECYAKVFDEFKPLV








EEPQNLIKQNCELFEQLGEYKFQNALL








VRYTKKVPQVSTPTLVEVSRNLGKVGS








KCCKHPEAKRMPCAEDYLSVVLNQLCV








LHEKTPVSDRVTKCCTESLVNRRPCFS








ALEVDETYVPKEFNAETFTFHADICTL








SEKERQIKKQTALVELVKHKPKATKEQ








LKAVMDDFAAFVEKCCKADDKETCFAE








EGKKLVAASQAALGLGGGGSGGGGSGG









GGSATISCRNEEGKAVDWFTFYKLPKR









QNKESGETGLEYLYLDSTTRSWRKSEQ








LMNDTKSVLGRTLQQLYEAYASKSNNT








AYLIYNDGVPKPVNYSRKYGHTKGLLL








WNRVQGFWLIHSIPQFPPIPEEGYDYP








PTGRRNGQSGICITFKYNQYEAIDSQL








LVCNPNVYSCSIPATFHQELIHMPQLC








TRASSSEIPGRLLTTLQSAQGQKFLHF








AKSDSFLDDIFAAWMAQRLKTHLLTET








WQRKRQELPSNCSLPYHVYNIKAIKLS








RHSYFSSYQDHAKWCISQKGTKNRWTC








IGDLNRSPHQAFRSGGFICTQNWQIYQ








AFQGLVLYYESCK








SEQ ID NO: 20




(Intentionally left blank)








SEQ ID NO: 21




(Intentionally left blank)








SEQ ID NO: 23




(Intentionally left blank)








SEQ ID NO: 24




(Intentionally left blank)








SEQ ID NO: 25




(Intentionally left blank)








SEQ ID NO: 26




(Intentionally left blank)








SEQ ID NO: 27




(Intentionally left blank)








SEQ ID NO: 28




(Intentionally left blank)








Human DNASE1L3 variants








SEQ ID NO: 29




(Intentionally left blank)








DNASE1L3, Q282_S305delinksK




(Signal Peptide; Mature Protein):




SEQ ID NO: 30





MSRELAPLLLLLLSIHSALAMRICSFN









VRSFGESKQEDKNAMDVIVKVIKRCDI








ILVMEIKDSNNRICPILMEKLNRNSRR








GITYNYVISSRLGRNTYKEQYAFLYKE








KLVSVKRSYHYHDYQDGDADVFSREPF








VVWFQSPHTAVKDFVIIPLHTTPETSV








KEIDELVEVYTDVKHRWKAENFIFMGD








FNAGCSYVPKKAWKNIRLRTDPRFVWL








IGDQEDTTVKKSINCAYDRIVLRGQEI








VSSVVPKSNSVFDFQKAYKLTEEEALD








VSDHFPVEFKLK








Murine DNase1L3 (O55070):




Amino acid sequence




(Signal Peptide; Mature Protein):




SEQ ID NO: 31





MSLHPASPRLASLLLFILALHDTLALR









LCSFNVRSFGASKKENHEAMDIIVKII








KRCDLILLMEIKDSSNNICPMLMEKLN








GNSRRSTTYNYVISSRLGRNTYKEQYA








FVYKEKLVSVKTKYHYHDYQDGDTDVF








SREPFVVWFHSPFTAVKDFVIVPLHTT








PETSVKEIDELVDVYTDVRSQWKTENF








IFMGDFNAGCSYVPKKAWQNIRLRTDP








KFVWLIGDQEDTTVKKSTSCAYDRIVL








CGQEIVNSVVPRSSGVFDFQKAYDLSE








EEALDVSDHFPVEFKLQSSRAFTNNRK








SVSLKKRKKGNRS








Rat DNase1L3 (O89107):




Amino acid sequence




(Signal Peptide; Mature Protein):




SEQ ID NO: 32





MSLYPASPYLASLLLFILALHGALSLR









LCSFNVRSFGESKKENHNAMDIIVKII








KRCDLILLMEIKDSNNNICPMLMEKLN








GNSRRSTTYNYVISSRLGRNTYKEQYA








FLYKEKLVSVKAKYLYHDYQDGDTDVF








SREPFVVWFQAPFTAAKDFVIVPLHTT








PETSVKEIDELADVYTDVRRRWKAENF








IFMGDFNAGCSYVPKKAWKNIRLRTDP








NFVWLIGDQEDTTVKKSTSCAYDRIVL








RGQEIVNSVVPRSSGVFDFQKAYELSE








EEALDVSDHFPVEFKLQSSRAFTNSRK








SVSLKKKKKGSRS








Chimpanzee DNase1L3 (H2QMU7):




Amino acid sequence




(Signal Peptide;




Mature Protein):




SEQ ID NO: 33





MSRELTPLLLLLLSIHSTLALRICSFN









VRSFGESKQEDQNAMDVIVKVIKRCDI








ILVMEIKDSNNRICPILMEKLNRNSRR








GITYNYVISSRLGRNTYKEQYAFLYKE








KLVSVKRSYHYHDYQDGDADVFSREPF








VVWFQSPHTAVKDFVIIPLHTTPETSV








KEIDELVEVYTDVKHRWKAENFIFMGD








FNAGCSYVPKKAWKNIRLRTDPRFVWL








IGDQEDTTVKKSINCAYDRIVLRGQEI








VSSVVPKSNSVFDFQKAYKLTEEEALD








VSDHFPVEFKLQSSRAFTNSKKSVTLR








KKTKSKRS








Olive baboon DNase1L3 (A0A2I3NFJ3):




Amino acid sequence




(Signal Peptide; Mature




Protein):




SEQ ID NO: 34





MSQELAPLLLLLLSIHSALALRICSFN









VRSFGESKQEDQNAMDVIVKVIKRCDI








MLLMEIKDSNNRICPVLMEKLNGNSRR








GIMYNYVISSRLGRNTYKEQYAFLYKE








KLVSVKRSYHYHDYQDGDVDVFSREPF








VVWFQSPHTAVKDFVIIPLHTTPETSV








KEIDELVDVYMDMKHRWKAENFIFMGD








FNAGCSYVPKKAWKNIRLRTDPRFVWL








IGDQEDTTVKRSTKCAYDRIVLRGQEI








VSSVVPKSNSVFDFQKAYKLTEEEALD








VSDHFPVEFKLQSSRAFTNSKKSVTVR








KKTKSKRS








Rabbit DNase1L3 (A0A2I3NFJ3):




Amino acid sequence




(Signal Peptide; Mature




Protein):




SEQ ID NO: 35





MSLGMSPASLLLLLLCLHGALALKLCS









FNVRSFGYSKRENRQAMDVIVKIIKRC








DIILLMEIKDSNNMICPTLMEKLNGNS








RRGITYNYVISSRLGRNVYKEQYAFLY








KEKLVTVKKNYLYHDYEAGDADAFSRE








PYVVWFQSPFTAVKDFVIVPLHTSPEA








SVKEIDELVDVYMDVKRRWNAENFIFM








GDFNAGCSYVPKKAWKNIRLRTDPRFV








WLIGDEEDTTVKKSTSCAYDRIVLRGQ








DIIRSVVPDSNGVFDFRKAYKLTEEEA








LDVSDHFPVEFKLQSSTAFTNSKKSVQ








PRKKAKAKRS








Dog DNase1L3 (F1P9C1):




Amino acid sequence (Signal




Peptide; Mature Protein):




SEQ ID NO: 36





MPRLPAFLLFLLLSISSALALRLCSFN









VRSFGGAKRENKNAMDVIVKVIKRCDI








ILLMEVKDSNNMICPTLLEKLNGNSRR








GIKYNYVISSRLGRNTYKEQYAFLYKE








KLVSVKKYYLYHDYQAGDADVFSREPF








VVWFQSPFTAVKDFVIVPLHTTPEASV








KEIDELVDVYLDVKRRWKAENFIFMGD








FNAGCSYVPKKAWKIIRLRTDPGFVWL








IGDQEDTTVKSSTHCAYDRIVLRGPEI








IRSVVPRSNSTFDFQKAFLLTEEEALN








VSDHFPVEFKLQSSRAFTNSKKSISPK








KKKVRHP








Pig DNase1L3 (A0A287B132):




Amino acid sequence (predicted




Signal Peptide; Mature Protein):




SEQ ID NO: 37





MSQLLVSLMLLLLSTHSSLALRICSFN









VRSFGESKKANCNAMDVIVKVIKRCDI








ILLMEIKDSNNMICPTLMEKLNGNSRR








SVTYNYVISSRLGRNTYKEQYAFLYKE








KLVSVKKSYLYHDYQSGDADVFSREPF








VVWFQSPYTAVKDFVIIPLHTTPETSV








KEIDELVDVYLDVKRRWEAENFIFMGD








FNAGCSYVPKKAWKNIRLRTDPMFIWL








IKDQEDTTVKKSINCAYDRIVLRGQEI








VSSVVPGSNSIFDFQKAYRLTEEKVRL








SFCLSVSPSGEDGVVSPRGIQATTGDT








LGHLTLSFKANDSLT








Guinea pig DNase1L3 (A0A286XK50):




Amino acid sequence




(Signal Peptide; Mature




Protein):




SEQ ID NO: 38





MSQTRPSLLLLLLAIHGALALKLCSFN









VRSFGESKKQNQNAMDVIVKIIKRCDL








MLLMEIKDSHNRICPMLMEKLNGNSRR








GTTYNYVISSRLGRNTYKEQYAFLYKE








KLVTVKDNYLFHDEDADVFSREPYVVW








FQSPHTAVKDFVIVPLHTTPETSVKEI








DELADVYTDVQRQWKVANFIFMGDFNA








GCSYVPKKAWKNIRLRTDPKFVWLIAD








DEDTTVKKSTSCAYDRIVLRGQEIVNS








VVPNSNGVFDFQKAYQLSEEQALEVSD








HFPVEFKLQSERAFTNNKKSVSLKKKK








KANRS








Cow DNase1L3 (F1MGQ1):




Amino acid sequence (Signal




Peptide; Mature Protein):




SEQ ID NO: 39





MPLPLACLLLLLLSTHSALALKICSFN









VRSFGESKKANCNAMDVIVKVIKRCDI








ILLMEIKDSSNRICPTLMEKLNGNSRK








GITYNYVISSRLGRNTYKEQYAFLYKE








KLVSVKQSYLYHDYQAGDADVFSREPF








VVWFQSPYTAVKDFVIVPLHTTPETSV








REIDELADVYTDVKRRWNAENFIFMGD








FNAGCSYVPKKAWKDIRLRTDPKFVWL








IGDQEDTTVKKSINCAYDRIVLRGQNI








VNSVVPQSNLVFDFQKAYRLSESKALD








VSDHFPVEFKLQSSRAFTNSKKSVSSK








KKKKTSHA








Elephant DNase1L3 (G3SXX1):




Amino acid sequence




(Signal Peptide; Mature Protein):




SEQ ID NO: 40




RSARMSQSLPALLLLLLLSVHGTLALR








VCSFNVRSFGETKRENQKVMDIIVKII








KRCDIMLLMEIKDSNNRICPMLLKRLN








GNSRRGIKYNYVISPRLGRNAYKEQYA








FLYMEKLLSVKKSYVYGDNQNGDADVF








SREPFVTWFQSPHTAVKDFVIVPLHTT








PETSIKEIDELVDVYMDVKKRWNAQNF








IFMGDFNAGCSYVPKKSWRNIRLRTDP








GFVWLIGDQEDTTVKESTNCAYDRIVL








RGQIISSVVPNSNSIFNFQKAYELSEE








EALNISDHFPVEFKLQSSRAIINSKKS








VSPKKKKKAKSS








LINKER SEQUENCES




SEQ ID NO: 41




GGGGS








SEQ ID NO: 42




GGGGSGGGGSGGGGS








SEQ ID NO: 43




APAPAPAPAPAPAP








SEQ ID NO: 44




AEAAAKEAAAKA








SEQ ID NO: 45




SGGSGSS








SEQ ID NO: 46




SGGSGGSGGSGGSGSS








SEQ ID NO: 47




SGGSGGSGGSGGSGGSGGSGGSGGSG








GSGS








SEQ ID NO: 48




GGSGGSGGSGGSGGSGGSGGSGGSGG








SGS








ACTIVATABLE LINKER SEQUENCES




FXIIa-susceptible linker




(Factor XI peptide):




SEQ ID NO: 49




CTTKIKPRIVGGTASVRGEWPWQVT








FXIIa-susceptible linker




(Prekallikrein peptide):




SEQ ID NO: 50




STRIVGG








FXIIa-susceptible linker




(Prekallikrein peptide):




SEQ ID NO: 51




VCTIKTSTRIVGGINSSWGEWPWQVS





Claims
  • 1. A method for treating a subject in need of extracellular DNA degradation, extracellular chromatin degradation, extracellular trap (ET) degradation and/or neutrophil extracellular trap (NET) degradation, the method comprising administering a therapeutically effective amount of a DNASE enzyme comprising an amino acid sequence that has at least 90% sequence identity to SEQ ID NO: 4, and having one or more amino acid substitutions of the C-terminal tail defined by SEQ ID NO: 10, and a C-terminal fusion to a carrier protein optionally through a linking sequence.
  • 2. The method of claim 1, wherein the DNASE enzyme has at least two amino acid substitutions in the C-terminal tail.
  • 3. The method of claim 1, wherein the DNASE enzyme has at least five amino acid substitutions in the C-terminal tail.
  • 4. The method of claim 1, wherein the DNASE enzyme has one or more amino acid substitutions of the C-terminal tail selected from: S282E, R285T, F287I, S290N, K291R, V294I, T295S, T295Q, L296V, L296P, L296S, R297K, K299R, T300K, T300A, S302G, S302A, S302V, S302T, K303N, K303S, K303R, R304H, R304S, S305P, S305T, and S305A.
  • 5. The method of claim 1, wherein the DNASE enzyme has one or more amino acid substitutions of the C-terminal tail selected from: (a) V294I;(b) T295S and T295Q;(c) L296V, L296P, and L296S;(d) R297K;(e) K299R;(f) T300K;(g) S302G, S302A, S302V, and S302T;(h) K303N, K303S, and K303R;(i) R304H and R304S; and(j) S305P, S305T, and S305A.
  • 6. The method of claim 4, wherein the DNASE enzyme has one or more amino acid substitutions of the C-terminal tail selected from K303N and K303S.
  • 7. The method of claim 1, further comprising the deletion of at least 3 amino acids of the C-terminal tail.
  • 8. The method of claim 7, wherein at least 5 amino acids of the C-terminal tail are deleted.
  • 9. The method of claim 7, wherein at least 12 amino acids of the C-terminal tail are deleted.
  • 10. The method of claim 1, wherein the carrier protein is albumin.
  • 11. The method of claim 1, wherein the carrier protein is an Fc domain.
  • 12. The method of claim 2, wherein the linking sequence is a flexible linker.
  • 13. The method of claim 12, wherein the linking sequence is composed predominately of Gly and/or Ser residues.
  • 14. The method of claim 2, wherein the linking sequence is a rigid linker.
  • 15. The method of claim 14, wherein the linking sequence comprises Pro or Pro-Ala motifs.
  • 16. The method of claim 14, wherein the linking sequence is an α-helical linker.
  • 17. The method of claim 1, wherein the DNASE enzyme exhibits protease resistance.
  • 18. The method of claim 1, wherein the DNASE enzyme is administered by parenteral administration.
  • 19. The method of claim 18, wherein the DNASE enzyme is administered by a route selected from intradermal, intramuscular, intraperitoneal, intraarticular, intravenous, and subcutaneous administration.
  • 20. The method of claim 1, wherein the subject has a chronic or acute inflammatory disorder.
  • 21. The method of claim 20, wherein the subject has an acute or chronic infection.
  • 22. The method of claim 1, wherein the subject has neutrophilia.
  • 23. The method of claim 1, wherein the subject has thrombosis or vascular occlusion.
  • 24. The method of claim 1, wherein the subject has an autoimmune disease.
  • 25. The method of claim 24, wherein the subject has systemic lupus erythematosus (SLE), lupus nephritis, rheumatoid arthritis, vasculitis, or systemic sclerosis.
  • 26. The method of claim 1, wherein the subject has diabetes mellitus.
  • 27. The method of claim 1, wherein the subject has sepsis.
  • 28. The method of claim 1, wherein the subject has an inflammatory disease of the respiratory tract.
  • 29. The method of claim 1, wherein the subject has a renal inflammatory disease, inflammatory disease related to transplated tissue, or cancer.
  • 30. The method of claim 1, wherein the subject has or is at risk of Nets occluding ductural systems.
PRIORITY

This Application is a continuation-in-part of U.S. application Ser. No. 17/427,974, filed Aug. 3, 2021, which is a national stage entry of PCT/US2020/016490, filed Feb. 4, 2020, and which claims the benefit of US Provisional Application No. 62/800,790, filed Feb. 4, 2019, each of which is hereby incorporated by reference in its entirety.

US Referenced Citations (25)
Number Name Date Kind
6482626 Baker et al. Nov 2002 B2
6656685 Utermohlen et al. Dec 2003 B2
7612032 Genkin et al. Nov 2009 B2
8388951 Genkin et al. Mar 2013 B2
8431123 Genkin et al. Apr 2013 B2
8535663 Genkin et al. Sep 2013 B2
8796004 Genkin et al. Aug 2014 B2
8916151 Genkin et al. Dec 2014 B2
9072733 Genkin et al. Jul 2015 B2
9149513 Bartoov et al. Oct 2015 B2
9198957 Ratner et al. Dec 2015 B2
9205133 Dawson et al. Dec 2015 B2
9248166 Gerkin et al. Feb 2016 B2
9402884 Burns Aug 2016 B2
9642822 Wagner May 2017 B2
9770492 Genkin et al. Sep 2017 B2
9845461 Genkin et al. Dec 2017 B2
9867871 Jain Jan 2018 B2
20040138156 Schneider et al. Jul 2004 A1
20090010966 Davis et al. Jan 2009 A1
20130149749 Holliger et al. Jun 2013 A1
20160251638 Posada et al. Sep 2016 A1
20160376366 Chang et al. Dec 2016 A1
20170196945 Wagner et al. Jul 2017 A1
20200024585 Fuchs et al. Jan 2020 A1
Foreign Referenced Citations (8)
Number Date Country
2011053982 May 2011 WO
2011131772 Oct 2011 WO
2018015474 Jan 2018 WO
2018064681 Apr 2018 WO
2018134403 Jul 2018 WO
2018134419 Jul 2018 WO
2019036719 Feb 2019 WO
2021142456 Jul 2021 WO
Non-Patent Literature Citations (35)
Entry
Andersen et al. 2014; Extending serum half-life of albumin by engineering neonatal Fe receptor (FcRn) binding. Journal of Biological Chemistry. 289(19): 13492-13502.
Napirei et al. 2009; Murine serum nucleases-contrasting effects of plasmin and heparin on the activities of DNase1 and DNase1-lie 3 (DNase113). FEBS Journal. 276: 1059-1073.
Shiokawa et al. 2003; Identification of two functional nuclear localization signals in DNase gamma and their roles in its apoptotic DNase activity. Biochem. J. 376: 377-381.
Berntsson et al., “Structural insight into DNA binding and oligomerization of the multifunctional Cox protein of bacteriophage P2”, Nucleic Acids Research, vol. 42, No. 4, 2014, pp. 2725-2735.
Hakkim et al., “Impairment of neutrophil extracellular trap degradation is associated with lupus nephritis”, PNAS, vol. 107, No. 21, 2010, pp. 9813-9818.
International Search Report and Written Opinion for International Application No. PCT/US2018/047084 , dated Feb. 15, 2019, 23 pages.
Keyel, “Dnases in health and disease”, Developmental Biology, vol. 429, 2017, pp. 1-11.
Kobayashi et al., “Synchronous Growth of Pichia Pastoris for a High-Rate Production of DNasel at Microquantities”, Department of Chemical Engineering. Toyko Institute of Technology. On-Line No. 833, 2004 pp. 1-6.
Perini et al., “Topical application of Acheflan on rat skin injury accelerates wound healing: a histopathological, immunohistochemical and biochemical study”, BMC Complementary and Alternative Medicine, 2015, vol. 15, No. 203, pp. 1-8.
Piccolo et al., “Intrapleural Tissue Plasminogen Activator and Deoxyribonuclease for Pleural Infection; An Effective and Safe Alternative to Surgery”, AnnalsATS, vol. 11, No. 9, 2014 , pp. 1419-1425.
Sisirak et al., “Digestion of Chromatin in Apoptotic Cell Microparticles Prevents Autoimmunity”, Cell vol. 166, 2016 , pp. 88-101.
Wilber et al., “Deoxyribonuclease I-like III Is an Inducible Macrophage Barrier to Liposomal Transfection”, MolecularTherapy, vol. 6, No. 1, 2002, pp. 35-42.
Jiménez-Alcázar et al., “Host DNases prevent vascular occlusion by neutrophil extracellular traps,” Science 358, pp. 1202-1206 (2017).
Branden et al., “Prediction, Engineering, and -Design of Protein Structures”, Introduction to Protein Structure, Garland Publishing Inc., New York, p. 247, 1991.
Seffernick, et al., “Melamine deaminase and Atrazine Chlorohydrolase: 98 Percent Identical but Functionally Different”, Journal Of Bacteriology, 2001, pp. 2405-2410.
Shiokawa et al., “Characterization of Human DNase I Family Endonucleases and Activation of DNase γ during Apoptosis”, Biochemistry 2001, 40, pp. 143-152.
Tang et al., “Identification of Dehalobacter Reductive Dehalogenases that Catalyse Dechlorination of Chloroform, 1,1,1-trichloroethane and 1,1-dichloroethane”, Phil Trans R Soc B, 368, 20120318, 1-10, 2013.
Witkowski et al., “Conversion of a β-Ketoacyl Synthase to a Malonyl Decarboxylase by Replacement of the Active-Site Cysteine with Glutamine”, Biochemistry 1999, 38, pp. 11643-11650.
Sadowski et al., “The sequence-structure relationship and protein function prediction”, Current Opinion in Structural Biology 19:357-362, 2009.
Ensembl ID No. ENSG00000163687, release 102, Nov. 2020.
Rodriguez et al., Gen Bank accession No. 013609 Sep. 27, 2017.
Baron et al., Cloning and characterization of an actin-resistant DNase I-like endonuclease secreted by macrophages, Gene, 1998, vol. 215 pp. 291-301.
Saito et al., Apoptotic DNA endonuclease (DNase-γ ) gene transfer induces cell death accompanying DNA fragmentation in human glioma cells, Journal of Neuro-Oncology, 2003, vol. 63, pp. 25-31.
Onuora, “DNASE1L3 prevents anti-DNA responses”, Nature Rev. Rheumatol., 2016, vol. 12 No. 437, 1 page.
Wang et al., “Targeting the extracellular scavenger DNASE1L3 on SLE”, J Xiangya Med, 2017, 3 pages.
Barnes et al. “Targeting potential drivers of COVID-19: Neutrophil extracellular traps”, J. Exp. Med., 2020, vol. 217, pp. 1-7.
Al-Mayouf et al., Loss-of-function variant in DNASE1L3 causes a familial form of systemic lupus erythematosus, Nature Genetics, 2011, vol. 43, No. 12, pp. 1186-1188.
Özçakar et al., DNASE1L3 Mutations in Hypocomplementemic Urticarial Vasculitis Syndrome, Arthritis & Rheumatism, 2013, vol. 65, No. 8, pp. 2183-2189.
Carbonella et al., An autosomal recessive DNASE1L3-related autoimmune disease with unusual clinical presentation mimicking systemic lupus erythematosus, Lupus, 2017, vol. 26, pp. 768-772.
Bruschi et al., Neutrophil extracellular traps (NET) induced by different stimuli: A comparative proteomic analysis, PLOS ONE, 2019, pp. 1-18.
Landhuis, “Spider-Man' Immune Response May Promote Severe COVID-19”, Sci. Am., 2020, pp. 1-7.
Reizis, “Project 3: The role of DNASE1L3 and its DNA substrate in lupus”, National Institute of Health (NIH), 2015, 5 pages.
Boettcher et al. “Therapeutic targeting of extracellular DNA improves the outcome of intestinal ischemic reperfusion injury in neonatal rats,” Scientific Reports, Nov. 13, 2017 (Nov. 13, 2017), vol. 7, No. 15377, pp. 1-10.
De Meyer et al. “Extracellular Chromatin Is an Important Mediator of Ischemic Stroke in Mice,” Arteriosclerosis, Thrombosis, and Vascular Biology, May 24, 2012 (May 24, 2012), vol. 32, No. 8, pp. 1884-1891.
International Search Report and Written Opinion for International Application No. PCT/US2020/016490, dated Jun. 23, 2020, 12 pages.
Related Publications (1)
Number Date Country
20220042002 A1 Feb 2022 US
Provisional Applications (1)
Number Date Country
62800790 Feb 2019 US
Continuation in Parts (1)
Number Date Country
Parent 17427974 US
Child 17509991 US