Increased polypeptide production yields of butyrylcholinesterase polypeptides for therapeutic use

Information

  • Patent Grant
  • 11473070
  • Patent Number
    11,473,070
  • Date Filed
    Monday, December 21, 2020
    3 years ago
  • Date Issued
    Tuesday, October 18, 2022
    a year ago
Abstract
The presently-disclosed subject matter describes fusion proteins comprising butyrylcholinesterase (BChE) having an improved production yield and biological half-life and nucleotides encoding the same.
Description
SEQUENCE LISTING

This application contains a sequence listing submitted in accordance with 37 C.F.R. 1.821, named 13177N 2396US ZHAN sequence listing.txt, created on Dec. 21, 2020, having a size of 67,608 bytes, which is incorporated herein by this reference.


TECHNICAL FIELD

The presently-disclosed subject matter relates to fusion proteins comprising butyrylcholinesterase (BChE) and having an improved production yield and biological half-life.


BACKGROUND

Human plasma butyrylcholinesterase (BChE) has a long history of clinical application, without any adverse events reported [1]. Two clinical trials (NCT00333515 and NCT00333528) of BChE protein were performed by Baxter Healthcare Corporation, showing that recombinant human BChE is also safe for use in humans.


It has been well known that BChE can intercept and destroy the organophosphorus (OP) nerve poisons before they reach their target—acetylcholinesterase (AChE) [1-3]. Thus, administration of BChE is recognized as an effective and safe medication for the prevention of organophosphorus (OP) nerve agent toxicity [3-6].


Because of the stoichiometric binding of BChE with OP nerve agent, a large amount of BChE protein is required to achieve its nerve protective effects in vivo. Thus, without an efficient BChE expression method, the clinical application of BChE is severely impeded by its actual availability, since the quantity of BChE protein purified from human plasma is very limited. Hence, it is highly desired to develop methods that can be used to efficiently produce BChE in a large-scale for further preclinical and clinical development.


Another driving force to solve this protein production problem comes from the potential application of mutant BChE for treatment of cocaine abuse. BChE is a major metabolic enzyme that catalyzes the hydrolysis of cocaine to produce biologically inactive metabolites. Unfortunately, the catalytic efficiency (kcat/KM) of wild-type BChE against naturally occurring (−)-cocaine is too low (kcat=4.1 min−1 and KM=4.5 μM) [7] to be effective for accelerating cocaine metabolism. Through structure and mechanism based computational design and wet experimental tests, a series of human BChE mutants with significantly improved catalytic efficiency against cocaine have been designed and discovered [7-12]. These BChE mutants have been recognized as true cocaine hydrolases (CocHs) in literature [8] when they have at least 1,000-fold improved catalytic efficiency against (−)-cocaine compared to wild-type human BChE [9-12].


The CocH-based approach has been recognized as a truly promising strategy for treatment of cocaine overdose and addiction [8, 13-15]. Thus, it is critical for further preclinical and clinical development towards the actual use of a BChE mutant in clinical practice to improve the protein production efficiency of the BChE and its mutants.


In fact, extensive efforts have been made to improve the protein production, with the goal to economically produce recombinant human BChE or BChE mutants. Expression in bacteria is recognized as the most economical method for producing recombinant proteins, but wild-type BChE expressed in bacteria cannot fold appropriately to become an active enzyme [16]. BChE proteins expressed in silkworm and insect cells were proven to be active [17, 18], but their pharmacokinetic profiles have not been characterized. Transgenic plants and animals were also generated to produce BChE or CocHs with a significantly improved efficiency, but the proteins produced usually have significantly shorter biological half-lives [19-22]. The short biological half-life is mainly explained by possibly incomplete post-translational modification causing the BChE or CocH to be taken up by asialo receptors in the liver [1].


Compared to all the expression systems above, CHO (Chinese-hamster ovary) cells provide more consistently proper protein post-translational modification [23]. Considering that the improper post-translational modification would not only shorten the protein's biological half-life, but also increase the risk of immunogenicity as an improper glycan structure might cause the protein to be recognized as an immunogen [1], CHO cells might be the most propriate system to produce the desirably safe and effective BChE (or BChE mutant) with a relatively long biological half-life. However, the biological half-life of the recombinant BChE or mutant produced in CHO [19-22, 24, 25] is still much shorter than that of native BChE. For example, CocH3 produced in CHO cells has a biological half-life of 7.3 hr in rats, which is considerably longer than that (˜13 min) of CocH3 expressed in plants [19, 24], but it is still much shorter than that (43 hr) of native BChE [26]. In addition, the low expression yield of BChE or its mutant in CHO cells is another major problem.


Thus, there remains a need in the art to efficiently produce active recombinant BChE and CocHs with a sufficiently long biological half-life


The presently disclosed subject matter identifies fusion proteins comprising BChE polypeptides that not only have a long biological half-life, but also a significantly-improved yield of protein production. Such polypeptides have utility in therapeutic treatment, for example, treatment of cocaine overdose and addiction, and treatment of OP detoxication.


SUMMARY

The presently-disclosed subject matter meets some or all of the above-identified needs, as will become evident to those of ordinary skill in the art after a study of information provided in this document.


This Summary describes several embodiments of the presently-disclosed subject matter, and in many cases lists variations and permutations of these embodiments. This Summary is merely exemplary of the numerous and varied embodiments. Mention of one or more representative features of a given embodiment is likewise exemplary. Such an embodiment can typically exist with or without the feature(s) mentioned; likewise, those features can be applied to other embodiments of the presently-disclosed subject matter, whether listed in this Summary or not. To avoid excessive repetition, this Summary does not list or suggest all possible combinations of such features.


The presently-disclosed subject matter includes fusion proteins comprising butyrylcholinesterase (BChE) and having an improved production yield and biological half-life, and methods for production of such fusion proteins.


One embodiment of the present invention is a polypeptide molecule, comprising: an Fc polypeptide joined to an N-terminal end of a butyrylcholinesterase (BChE) polypeptide. In other embodiments of the present invention, an Fc polypeptide is joined to a C-terminal end of a butyrylcholinesterase (BChE) polypeptide. In some embodiments of the present invention, the Fc polypeptide is optionally joined to the BChE polypeptide via a linker, the linker comprising a sequence selected from the sequences of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 19, SEQ ID NO: 36, and SEQ ID NO: 37. In certain embodiments of the present invention, the Fc polypeptide has the sequence of SEQ ID NO: 8, or a fragment thereof, wherein the Fc polypeptide or fragment thereof includes 3 to 8 amino acid substitutions at 3 to 8 of residues selected from 1, 6, 12, 15, 24, 38, 40, 42, 58, 69, 80, 98, 101, 142, and 144. In further embodiments of the present invention, the Fc polypeptide is a fragment wherein 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16 amino acids are removed from the N-terminus of SEQ ID NO: 8. In some embodiments, the Fc polypeptide includes mutations as set forth in Table A, relative to SEQ ID NO: 8.









TABLE A







Substitutions relative to SEQ ID NO: 8 for exemplary Fc Polypeptides






















Fc
1
6
12
15
24
38
40
42
58
69
80
98
101
142
144





M3
A1V












D124E
L144M


M8
A1V







E58Q
E69Q
E80Q
D98N
N101D
D124E
L144M


M5
A1Q
C6S
C12S
C15S
P24S












M4
A1V




M38Y







D142E
L144M


M4′
A1V






T42E





D142E
L144M


M5′
A1V




M38Y
S40T






D142E
L144M


M6
A1V




M38Y
S40T
T42E





D142E
L144M


M6′
A1Q
C6S
C12S
C15S
P24S
M38Y











M7
A1Q
C6S
C12S
C15S
P24S
M38Y
S40T










M8′
A1Q
C6S
C12S
C15S
P24S
M38Y
S40T
T42E









In some embodiments, the BChE polypeptide is an BChE polypeptide fragment that further includes amino acid substitutions as set forth in Table B, relative to SEQ ID NO: 10.









TABLE B







Substitutions relative to SEQ ID NO: 10 for exemplary


BChE Polypeptides














199
227
285
286
287
328
332
441





A199S




A328W
Y332G



A199S
F227A



A328W
Y332G



A199S



S287G
A328W
Y332G



A199S
F227A


S287G
A328W
Y332G



A199S
F227A


S287G
A328W

E441D


A199S
F227A
P285A
S287G
A328W
Y332G




A199S
F227A
P285S
S287G
A328W
Y332G




A199S
F227A
P285Q
S287G
A328W
Y332G




A199S
F227P


S287G
A328W
Y332G



A199S
F227A


S287G
A328W
Y332G



A199S
F227A

L286M
S287G
A328W
Y332G



A199S

P285Q

S287G
A328W
Y332G



A199S

P285I

S287G
A328W
Y332G



A199S
F227G


S287G
A328W
Y332G



A199S

P285S

S287G
A328W
Y332G



A199S
F227V


S287G
A328W
Y332G



A199S

P285G

S287G
A328W
Y332G



A199S
F227I


S287G
A328W
Y332G



A199S
F227L


S287G
A328W
Y332G



A199S

P286M
L286M
S287G
A328W
Y332G



A199S
F227A


S287G
A328W
Y332G



A199S
F227S


S287G
A328W
Y332G



A199S
F227T


S287G
A328W
Y332G



A199S
F227M


S287G
A328W
Y332G



A199S
F227C


S287G
A328W
Y332G



A199S
F227A
P285N

S287G
A328W
Y332G



A199S
F227P
P285A

S287G
A328W
Y332G



A199S
F227S
P285Q

S287G
A328W
Y332G



A199S
F227S
P285S

S287G
A328W
Y332G



A199S
F227S
P285G

S287G
A328W
Y332G



A199S
F227P
P285S
L286M
S287G
A328W
Y332G



A199S
F227A
P285S

S287G
A328W

E441D


A199S
F227A
P285A

S287G
A328W

E441D


A199S
F227P

L286M
S287G
A328W
Y332G



A199S
F227G
P285A

S287G
A328W
Y332G



A199S
F227G
P285G

S287G
A328W
Y332G



A199S
F227G
P285Q

S287G
A328W
Y332G



A199S
F227G
P285S

S287G
A328W
Y332G



A199S
F227A
P285E

S287G
A328W
Y332G



A199S
F227P
P285N

S287G
A328W
Y332G



A199S
F227S
P285A

S287G
A328W
Y332G



A199S
F227S
P285N

S287G
A328W
Y332G



A199S
F227S

L286M
S287G
A328W
Y332G



A199S
F227G

L286M
S287G
A328W
Y332G










In some embodiments of the present invention, the Fc polypeptide is selected from SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 28, SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 31, SEQ ID NO: 32, SEQ ID NO: 33, and SEQ ID NO: 34. In further embodiments, the BChE polypeptide has the sequence of SEQ ID NO: 10 or a fragment thereof, wherein the BChE polypeptide or fragment thereof includes 3 to 8 amino acid substitutions at 3 to 8 of residues chosen from 199, 227, 285, 286, 287, 328, 332, and 441. In other embodiments, the BChE polypeptide has a group of amino acid substitutions selected from A199S, F227A, F227S, F227Q, F227I, F227G, F227V, F227I, F227L, F227S, F227T, F227M, F227C, P285A, P285S, P285Q, P285I, P285G, P285M, P285N, P285E, S287G, A328W, Y332G, E441D, and combinations thereof. In other embodiments of the presently disclosed matter, the BChE polypeptide is a fragment wherein from 1 to 116 amino acids are removed from the N-terminus of SEQ ID NO: 10. In some embodiments of the invention, the BChE polypeptide is a fragment wherein from 1 to 432 amino acids are removed from the C-terminus of SEQ ID NO: 10. In some embodiments, the BChE polypeptide has a group of amino acid substitutions selected from A199S, F227A, F227S, F227Q, F227I, F227G, F227V, F227I, F227L, F227S, F227T, F227M, F227C, P285A, P285S, P285Q, P285I, P285G, P285M, P285N, P285E, S287G, A328W, Y332G, E441D, and combinations thereof. In other embodiments of the present invention, the transient expression level of the polypeptide is at least about 9 times higher than a reference BChE polypeptide that does not include the Fc polypeptide and linker. In some embodiments of the present invention, the polypeptide molecule is the polypeptide of SEQ ID NO: 35. In other embodiments, the BChE polypeptide is selected from: SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, and SEQ ID NO: 15. In further embodiments, Fc polypeptide is selected from SEQ ID NO: 16, SEQ ID NO: 17, and SEQ ID NO: 18.


The presently-disclosed subject matter also relates to a nucleotide molecule, comprising: a nucleotide encoding an Fc polypeptide joined by a nucleotide encoding a linker to a 5′ end of a nucleotide encoding a butyrylcholinesterase (BChE) polypeptide. In some embodiments, the nucleotide encoding the linker comprises a sequence chosen from the sequences of SEQ ID NO: 1, SEQ ID NO: 3, and SEQ ID NO: 5. In some embodiments, the nucleotide encoding the Fc polypeptide has the sequence of SEQ ID NO: 7 or a fragment thereof, wherein the Fc polypeptide or fragment thereof includes 3 to 8 amino acid substitutions at 3 to 8 of residues chosen from 1, 6, 12, 15, 24, 38, 40, 42, 58, 69, 80, 98, 101, 142, and 144 relative to SEQ ID NO: 8. In further embodiments, the nucleotide encoding the BChE polypeptide has the sequence of SEQ ID NO: 9 or a fragment thereof, wherein the BChE polypeptide or fragment thereof includes 3 to 8 amino acid substitutions at 3 to 8 of residues chosen from 199, 227, 285, 286, 287, 328, 332, and 441 relative to SEQ ID NO: 10. In some embodiments of the present invention, the nucleotide molecule is within an expression vector.


The present invention also relates to a method of producing a polypeptide molecule including a BChE polypeptide, comprising: (a) providing in a vector a nucleotide sequence chosen from (i) a nucleotide sequence encoding the polypeptide molecule of claim 1, or (ii) a nucleotide sequence of claim 14; and (b) transfecting cells with the vector and allowing the cells to express the polypeptide molecule; and (c) isolating the polypeptide molecule. In further embodiments, there is at least about a 9-fold improvement in the yield of expression of the polypeptide molecule as compared to expression of a reference BChE polypeptide.





BRIEF DESCRIPTION OF THE DRAWINGS

The presently-disclosed subject matter will be better understood, and features, aspects and advantages other than those set forth above will become apparent when consideration is given to the following detailed description thereof. Such detailed description makes reference to the following drawings, wherein:



FIG. 1 includes a schematic illustrating four Fc-fused protein with various linkers (E1 with SEQ ID NO: 19; E2 with SEQ ID NO: 4; E3 with SEQ ID NO: 6; and E4 with SEQ ID NO: 2), as well as a non-linked Fc-BChE. The small grey box represents an IL-2 secretion signal peptide; the black box represents the sequence of a Fc polypeptide, as disclosed herein; and the white box represents the sequence of a BChE polypeptide, as disclosed herein.



FIG. 2 includes a Coomassie-Blue stained native electrophoresis gel of purified fusion proteins, showing that the native structures of all fusion proteins exist in a dimer.



FIG. 3 includes kinetic data obtained in vitro for (−)-cocaine hydrolysis catalyzed by the fusion proteins. The reaction rate is represented in μM min′ per nM enzyme.



FIG. 4 is a graph illustrating the time-dependent concentration of Fc(M3)-(PAPAP)2-CocH3 (SEQ ID NO: 16-SEQ ID NO: 2-SEQ ID NO: 15) fusion. in the plasma of rats after IV administration of the enzyme (0.075 mg/kg) determined in triplicate.



FIG. 5 shows Fc fusion protein CocH-LAF6 plots of the measured protein binding (%) vs the Fc-fused protein concentration.





While the disclosure is susceptible to various modifications and alternative forms, specific embodiments thereof have been shown by way of example in the drawings and are herein described below in detail. It should be understood, however, that the description of specific embodiments is not intended to limit the disclosure to cover all modifications, equivalents and alternatives falling within the spirit and scope of the disclosure as defined by the appended claims.


DESCRIPTION OF EXEMPLARY EMBODIMENTS

The details of one or more embodiments of the presently-disclosed subject matter are set forth in this document. Modifications to embodiments described in this document, and other embodiments, will be evident to those of ordinary skill in the art after a study of the information provided in this document. The information provided in this document, and particularly the specific details of the described exemplary embodiments, is provided primarily for clearness of understanding and no unnecessary limitations are to be understood therefrom. In case of conflict, the specification of this document, including definitions, will control.


While the terms used herein are believed to be well understood by those of ordinary skill in the art, certain definitions are set forth to facilitate explanation of the presently-disclosed subject matter.


Unless defined otherwise, all technical and scientific terms used herein have the same meaning as is commonly understood by one of skill in the art to which the invention(s) belong.


All patents, patent applications, published applications and publications, GenBank sequences, databases, websites and other published materials referred to throughout the entire disclosure herein, unless noted otherwise, are incorporated by reference in their entirety.


Where reference is made to a URL or other such identifier or address, it understood that such identifiers can change and particular information on the internet can come and go, but equivalent information can be found by searching the internet. Reference thereto evidences the availability and public dissemination of such information.


As used herein, the abbreviations for any protective groups, amino acids and other compounds, are, unless indicated otherwise, in accord with their common usage, recognized abbreviations, or the IUPAC-IUB Commission on Biochemical Nomenclature (see, Biochem. (1972) 11(9):1726-1732).


Although any methods, devices, and materials similar or equivalent to those described herein can be used in the practice or testing of the presently-disclosed subject matter, representative methods, devices, and materials are described herein.


Following long-standing patent law convention, the terms “a”, “an”, and “the” refer to “one or more” when used in this application, including the claims. Thus, for example, reference to “a biomarker” includes a plurality of such biomarkers, and so forth.


Unless otherwise indicated, all numbers expressing quantities of ingredients, properties such as reaction conditions, and so forth used in the specification and claims are to be understood as being modified in all instances by the term “about”. Accordingly, unless indicated to the contrary, the numerical parameters set forth in this specification and claims are approximations that can vary depending upon the desired properties sought to be obtained by the presently-disclosed subject matter.


As used herein, the term “about,” when referring to a value or to an amount of mass, weight, time, volume, width, length, height, concentration or percentage is meant to encompass variations of in some embodiments ±10%, in some embodiments ±5%, in some embodiments ±1%, in some embodiments ±0.5%, and in some embodiments ±0.1% from the specified amount, as such variations are appropriate to perform the disclosed method.


As used herein, ranges can be expressed as from “about” one particular value, and/or to “about” another particular value. It is also understood that there are a number of values disclosed herein, and that each value is also herein disclosed as “about” that particular value in addition to the value itself. For example, if the value “10” is disclosed, then “about 10” is also disclosed. It is also understood that each unit between two particular units are also disclosed. For example, if 10 and 15 are disclosed, then 11, 12, 13, and 14 are also disclosed.


As used herein, “optional” or “optionally” means that the subsequently described event or circumstance does or does not occur and that the description includes instances where said event or circumstance occurs and instances where it does not. For example, an optionally variant portion means that the portion is variant or non-variant.


As used herein, the term “subject” refers to a target of administration. The subject of the herein disclosed methods can be a mammal. Thus, the subject of the herein disclosed methods can be a human, non-human primate, horse, pig, rabbit, dog, sheep, goat, cow, cat, guinea pig or rodent. The term does not denote a particular age or sex. Thus, adult and newborn subjects, as well as fetuses, whether male or female, are intended to be covered. A “patient” refers to a subject afflicted with a disease or disorder. The term “patient” includes human and veterinary subjects.


As used herein, the term “BChE polypeptide” can refer to various mutations and truncations of the BChE protein including the mutations that are characterized by cocaine hydrolase (CoCH). BChE polypeptide for example includes, but it not limited to, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, and SEQ ID NO: 15.


EXAMPLES

The fusion proteins as disclosed herein were designed in view of a number of considerations. For example, they make use of a protein that is normally expressed in a high level as the N-terminal fusion partner to improve the expression of protein of interest [27, 28]. It was contemplated that the N-terminal fusion partner could “fool” the cellular process into expressing the fusion protein at a high level [28]. For another example, human IgG has a very long biological half-life (t1/2). The fragment crystallizable (Fc) region of IgG binds to the neonatal Fc receptor (FcRn) in the acidic environment of the endosome and later is transported to the cell surface where, upon exposure to a neutral pH, IgG is released back into the main bloodstream [29]. In addition, IgG is the most common type of antibody found in the circulation, and can be expressed in CHO cells with a yield of more than 1 g/L [30].


The present inventors sought to design a long-acting CocH form which has not only a prolonged biological half-life without affecting the catalytic activity, but also an improved expression level in CHO cells. For this purpose, exemplary embodiments were prepared for testing, starting from CocH3 (the A199S/F227A/S287G/A328W/Y332G mutant [9] of human BChE) (SEQ ID NO: 12), a IL-2 signal peptide followed by Fc(M3) (the A1V/D142E/L144M mutant [31] of Fc)(SEQ ID NO: 16), which was fused with the N-terminal of CocH3 (SEQ ID NO: 12). Then the tetramerization domain (amino-acid residues 530 to 574) of CocH3 was deleted to minimize the possibility of affecting the correct folding of Fc(M3) or CocH3. On the other hand, it was contemplated that the presence of Fc(M3) might break the tetramer structure, resulting in a long and flexible peptide, which could be proteolyzed easily. In addition, according to computational modeling (data not shown), directly fusing Fc(M3) with the N-terminal of CocH3 could affect the entrance of substrate to the active site of CocH3, thus affecting the catalytic activity of CocH3. Hence, several types of linkers were selected and inserted between Fc(M3) and CocH3. In this way, various Fc(M3)-linker-CocH3 entities were prepared and tested for their catalytic activity against cocaine, protein expression yields in CHO cells, and pharmacokinetic profile (for the most promising entity), leading to identification of a promising Fc(M3)-linker-CocH3 entity, as discussed below.


Materials and Methods


Materials


Q5® Site-Directed Mutagenesis Kit was ordered from New England Biolabs (Ipswich, Mass.). All oligonucleotides were synthesized by Eurofins MWG Operon (Huntsville, Ala.). Chinese Hamster Ovary-suspension (CHO-S) cells, FreeStyle™ CHO Expression Medium, Fetal Bovine Serum (FBS), 4-12% Tris-Glycine Mini Protein Gel, and SimpleBlue SafeStain were obtained from Invitrogen (Grand Island, N.Y.). TransIT-PRO® Transfection Kit was purchased from Minis (Madison, Wis.). The rmp Protein A Sepharose Fast Flow was from GE Healthcare Life Sciences (Pittsburgh, Pa.). (−)-Cocaine was provided by the National Institute on Drug Abuse (NIDA) Drug Supply Program (Bethesda, Md.); and [3H](−)-Cocaine (50 Ci/mmol) was obtained from PerkinElmer (Waltham, Mass.). All other materials were from Sigma-Aldrich (St Louis, Mo.) or Thermo Fisher Scientific (Waltham, Mass.).


Preparation of Gene Fusion Constructs in pCMV-MCS


Q5® Site-Directed Mutagenesis Kit was used to introduce each linker between Fc(M3) and CocH3. The pCMV-Fc(M3)-CocH3, constructed in a previous study [32] to encode N-terminal Fc-fused CocH3 without a linker, was used as the template. PCR reactions with Q5 hot start high-fidelity DNA polymerase along with primers listed in Table 1 were utilized to create insertions. Then 1 μl of each PCR product was incubated with Kinase-Ligase-DpnI enzyme mix for 15 minutes at room temperature. These steps allowed for rapid circulation of the PCR product and removal of the template DNA. 5 μl of final product was added to 50 μl of chemically-competent E. coli cells for transformation. All obtained plasmid encoding different Fc-fused CocH3 were confirmed by DNA sequencing.









TABLE 1







Examples of primers for inserting various linkers









Linker
Primer name
Primer sequence





EAAAK
EAAAK-F
5′-G TCT CCG GGT AAA GAG GCT GCC GCC




AAG GAA GAT GAC ATC A-3′ (SEQ ID NO:




20)



EAAAK-R
5′-CTT GGC GGC AGC CTC TTT ACC CGG




AGA CAG GGA GAG-3′ (SEQ ID NO: 21)





PAPAP
PAPAP-F
5′-G TCT CCG GGT AAA CCT GCT CCA GCC




CCG GAA GAT GAC ATC A-3′ (SEQ ID NO:




22)



PAPAP-R
5′-CGG GGC TGG AGC AGG TTT ACC CGG




AGA CAG GGA GAG-3′ (SEQ ID NO: 23)





GGGSGGGS
(G3S)2-F
5′-G TCT CCG GGT AAA GGT GGA GGT TCC




GGT GGA GGT TCC GAA GAT GAC ATC A-3′




(SEQ ID NO: 24)



(G3S)2-R
5′- GGA ACC TCC ACC GGA ACC TCC ACC




TTT ACC CGG AGA CAG GGA GAG-3′ (SEQ




ID NO: 25)





PAPAPPAPAP
(PAPAP)2-F
5′-G TCT CCG GGT AAA CCT GCT CCA GCC




CCG CCT GCT CCA GCC CCG GAA GAT GAC




ATC A-3′ (SEQ ID NO: 26)



(PAPAP)2-R
5′- CGG GGC TGG AGC AGG CGG GGC TGG




AGC AGG TTT ACC CGG AGA CAG GGA




GAG-3′ (SEQ ID NO: 27)









Expression and Purification


CHO-S cells were grown under the condition of 37° C. and 8% CO2 in a humidified atmosphere. Once cells grown to a density of ˜1.0×106 cells/ml, cells were transfected with plasmids encoding various proteins using TransIT-PRO® transfection kit. The culture medium was harvested 7 days after transfection. Enzyme secreted in the culture medium was purified by protein A affinity chromatography described previously [31]. Briefly, pre-equilibrated rmp Protein A Sepharose Fast Flow was mixed with cell-free medium, and incubated overnight at 6° C. with occasional stirring. Then the suspension was packed in a column, washed with 20 mM Tris.HCl (pH 7.4), and eluted by adjustment of salt concentration and pH. The eluate was concentrated and dialyzed in storage buffer (50 mM HEPES, 20% sorbitol, 1 M glycine, pH 7.4). Purified proteins were analyzed by native PAGE electrophoresis.


In Vitro Activity Assay Against (−)-Cocaine.


A radiometric assay based on toluene extraction of [3H](−)-cocaine labeled on its benzene ring was used to determine the catalytic activity of proteins [9, 11, 33]. Reactions were initiated by adding 150 μl enzyme solution (100 mM phosphate buffer, pH 7.4) to 50 μl [3H](−)-cocaine solution with varying concentration. Then 200 μl of 0.1 M HCl was added to stop each reaction and neutralize the liberated benzoic acid while ensuring a positive charge on the residual (−)-cocaine. [3H]Benzoic acid was extracted by 1 ml of toluene and measured by scintillation counting. Catalytic rate constant (kcat) and Michaelis-Menten constant (Km) were determined by fitting the substrate concentration-dependent data using Michaelis-Menten kinetics.


Determination of Relative Expression Level of Proteins


Cells were grown in 12-well plates to a density of ˜1.0×106 cells/ml. Then cells were transfected with plasmids encoding different proteins using the same method described above. The test was tripled for each protein, occupying 3 out of 12 wells in a plate. Medium was collected from each well 3 days post the transfection. Cells were removed by centrifuge at 4000 rpm for 15 min, and the catalytic activity of each sample against cocaine was determined using radiometric assay described above. Protein concentration was calculated by dividing the catalytic activity by the kcat (determined by using the aforementioned purified protein) for each specific protein.


Determination of Biological Half-Life in Rats


Male Sprague-Darley rats (220-250 g) were ordered from Harlan (Harlan, Indianapolis, Ind.), and housed initially as one or two rats per cage. All rats were allowed ad libitum access to food and water and maintained on a 12 h light/12 h dark cycle, with the lights on at 8:00 a.m. at a room temperature of 21-22° C. Experiments were performed in a same colony room in accordance with the Guide for the Care and Use of Laboratory Animals as adopted and promulgated by the National Institutes of Health. The animal procedure was approved by the IACUC (Institutional Animal Care and Use Committee) as part of the animal protocol 2010-0722 on Jun. 21, 2016 at the University of Kentucky. Rats were injected with the purified Fc(M3)-(PAPAP)2—CocH3 protein via tail vein (0.075 mg/kg). Blood samples were collected from saphenous vein puncture. Approximately 100 μl blood was collected by using heparin-treated capillary tube at various time points after enzyme administration. Collected samples were centrifuged for 15 min at a speed of 5000 g to separate the plasma, which was kept at 4° C. before analysis. Radiometric assay using 100 μM (−)-cocaine was carried out to measure the active enzyme concentration in plasma.


Results and Discussion


Optimization of Fc-Fused CocH3 Entity with a Linker


Four different linkers, including flexible linkers (GGGSGGGS) (SEQ ID NO: 6) (GGGGSGGGGS)(SEQ ID NO: 36), (GGGGGGSGGGGGGS)(SEQ ID NO: 37) and three rigid linkers (EAAAK—SEQ ID NO: 19), (PAPAP—SEQ ID NO: 4), and (PAPAPPAPAP-SEQ ID NO: 2), were utilized in this study. Previous studies reported in literature indicated that a linker similar to these could separate carrier protein and functional protein effectively and lead to improved biological activity of the fusion proteins with a linker. The four fusion proteins (see FIG. 1) with various linkers were then expressed, purified, and characterized for their catalytic activity against cocaine by performing the sensitive radiometric assay using [3H](−)-cocaine with various concentrations. All of the fusion proteins have a size ˜210 kDa (FIG. 2), which is consistent with the expected dimeric structure with disulfide bonds formed on the Fc part. CocH3, which was produced predominantly in a tetramer form in a previous study [9] has a kcat of 5700 min−1; however, the fusion protein, Fc(M3)-CocH3, which was constructed without any linker in a previous study [32], exhibited only ˜30% catalytic activity against cocaine as compared to the corresponding unfused CocH3.


To optimize the construct of Fc(M3)-CocH3, the above mentioned four linkers were used to eliminate the negative effects of N-terminal Fc portion on the catalytic activity of C-terminal CocH3. As seen in Table 2 and FIG. 3, with the insertion of a flexible linker (G3S)2, CocH3(529)-(G3S)2-Fc(M3) [SEQ ID NO: 13-SEQ ID NO: 6-SEQ ID NO: 16] has a kcat of 3579 min−1 which was 2-fold higher than that of the fusion protein without a linker. However, the kcat value of CocH3(529)-(G3S)2-Fc(M3) was still lower than that of the corresponding unfused CocH3 (SEQ ID NO: 13) (3579 min−1 compared to 5700 min−1). Nevertheless, Fc(M3)-EAAAK-CocH3[SEQ ID NO: 16-SEQ ID NO: 19-SEQ ID NO: 13], Fc(M3)-PAPAP-CocH3[SEQ ID NO: 16-SEQ ID NO: 4-SEQ ID NO: 13], and Fc(M3)-(PAPAP)2-CocH3[SEQ ID NO: 16-SEQ ID NO: 2-SEQ ID NO: 13] all have a catalytic activity comparable to the kcat of the unfused CocH3. The kcat values of Fc(M3)-EAAAK-CocH3, Fc(M3)-PAPAP-CocH3, and Fc(M3)-(PAPAP)2—CocH3 are 5287, 6078, and 5270 min−1, respectively. As a rigid linker used in these fusion proteins provides enough space between the Fc(M3) and CocH3 domains, it is likely that these two protein domains can be fold correctly without affecting each other in terms of their functions, including the entrance to active site of CocH3 for cocaine. Further longer linker were not tested in this study for a couple of reasons. First, the currently designed linker could provide enough space for the two protein domains, thus fusing the Fc(M3) is not expected to affect the catalytic activity of CocH3 against cocaine. Further, a further longer linker between CocH3 and Fc could be proteolyzed easily and, thus, could shorten the biological half-life of the fusion protein [3].









TABLE 2







Kinetic parameters determined for (-)-cocaine hydrolysis catalysed


by the fusion proteins











Protein



kcat/KM


#
Enzyme
Km (μM)
kcat (min-1)
(min-1 M-1)





1*
Fc(M3)CocH3(529)
3.9 ± 0.5
1835 ± 61
4.7 × 108



(SEQ ID NO: 16-SEQ ID NO:






13)








2*
Fc(M3)-G6S-CocH3
4.4 ± 0.6
5694 ± 238
1.3 × 109



(SEQ ID NO: 16-SEQ ID NO:






37-SEQ ID NO: 13)








3
Fc(M3)-EAAAK-CocH3(529)
4.3 ± 0.4
5684 ± 148
1.3 × 109



(SEQ ID NO: 16-SEQ ID NO:






19-SEQ ID NO: 13)








4
Fc(M3)-PAPAP-CocH3(529)
4.2 ± 0.5
6078 ± 202
1.4 × 109



(SEQ ID NO: 16-SEQ ID NO:






4-SEQ ID NO: 13)








5
Fc(M3)-(G3S)2-CocH3(529)
3.7 ± 0.4
3579 ± 104
9.6 × 108



(SEQ ID NO: 16-SEQ ID NO:






6-SEQ ID NO: 6-SEQ ID NO:






13)








6
Fc(M3)-(PAPAP)2-CocH3(529)
4.5 ± 0.5
5666 ± 148
1.3 × 109



(SEQ ID NO: 16-SEQ ID NO:






2-SEQ ID NO: 13)





*The kcat and KM of the enzymes against (-)-cocaine were reported in ref. [32]






Effects of the Linker on the Expression of Fc(M3)-Fused CocH3 Protein


Fc(M3)-EAAAK-CocH3, Fc(M3)-PAPAP-CocH3, and Fc(M3)-(PAPAP)2—CocH3 were further expressed together with Fc(M3)-CocH3 and the unfused CocH3 for comparison of relevant protein expression levels. All five proteins were expressed in the same plate under the same conditions at the same time. Cells in each well transfected using the same method and cultured under the same conditions after the transfection. All media were collected 3 days after the transfection. Protein expression level in each well was determined using the radiometric assay using 100 μM [3H](−)-cocaine. As seen in Table 3, the expression of the unfused CocH3 was 0.5 mg/L 3 days after the transient transfection. Usually, inserting the Fc portion at the N-terminal of the target protein could significantly improve the protein expression level. In this study, directly fusing Fc to the N-terminal of CocH3 increased the protein expression level by ˜2-fold. However, as Fc(M3)-CocH3 protein had only ˜30% catalytic activity against cocaine as compared to the unfused CocH3 [32]. As Fc(M3) domain sterically interferes with the CocH3 domain activity and lowers its catalytic activity against cocaine, it is also possible that this steric interference affects the efficiency of the protein folding. Therefore, an appropriate linker capable of avoiding such steric interference may not only improve the catalytic activity against cocaine, but also increase the protein expression level. As shown in Table 3, the protein expression yields of Fc(M3)-EAAAK-CocH3 and Fc(M3)-PAPAP-CocH3 was 4.8, and 5.2 mg/L, respectively. Linkers EAAAK, and PAPAP improved the yield of Fc(M3)-CocH3 protein expression by ˜9 and ˜10 folds, respectively. Among all fusion proteins constructed in this study, Fc(M3)-(PAPAP)2—CocH3 has the highest protein expression yield. The linker (PAPAPPAPAP) increased the yield of protein expression by ˜10 fold compared to the corresponding fusion protein without a linker. Further, compared to the corresponding unfused protein (CocH3), Fc(M3)-(PAPAP)2-CocH3 had a ˜21-fold improved yield of protein expression.









TABLE 3







Transient expression levels of fusion proteins, in


comparison of CocH3










Expression



Enzyme
level (mg/L)
Ratio





CocH3 (SEQ ID NO: 13)
 0.5 ± 0.1
 1





Fc(M3)-CocH3 (SEQ ID NO:
 1.1 ± 0.2
 2


16-SEQ ID NO: 13)







Fc(M3)-EAAAK-CocH3
 4.8 ± 0.6
 9


(SEQ ID NO: 16-SEQ ID NO:




19-SEQ ID NO: 13)







Fc(M3)-PAPAP-CocH3
 5.2 ± 0.6
10


(SEQ ID NO: 16-SEQ ID NO:




4-SEQ ID NO: 13)







Fc(M3)-(PAPAP)2-CocH3
10.9 ± 1.1
21


(SEQ ID NO: 16-SEQ ID NO:




2-SEQ ID NO: 13)









It should be pointed out that the transient expression method (with the protein expression within only three days) in this study was used only for the purpose of comparing the relative expression levels of various fusion proteins and unfused protein under the same conditions. So, the key results of this study are the relative protein expression levels, rather than the absolute protein expression levels. The absolute protein expression levels are expected to significantly increase when the stable CHO cell lines are developed and used to express the same proteins; of course, development of a stable cell line is a very time-consuming process. For example, using a lentivirus-based repeated-transduction method which was established in a previous study [24], the protein expression yield of the unfused CocH3 reached ˜10 mg/L in a flask-based culture. Thus, one would reasonably expect that an appropriately developed stable CHO cell line might be able to express ˜200 mg/L Fc(M3)-(PAPAP)2-CocH3 protein by using the same lentivirus-based repeated-transduction method. The protein expression yield could be improved further by optimizing of the culture conditions, such as cell density, medium, and culture temperature.


Biological Half-Life of Fc(M3)-(PAPAP)2—CocH3 in Rats


Pharmacokinetic testing was carried out to determine biological half-life of Fc(M3)-(PAPAP)2-CocH3. Rats (n=3) were administered IV with 0.075 mg/kg of the purified protein. The blood was collected at 1 hr, 4 hr, 8 hr, 12 hr, 1 day and once each day within 14 days after the enzyme injection. Depicted in FIG. 4 is the time course of the percentage of the enzyme activity remained after enzyme injection. The time-dependent data were fitted to a double-exponential equation ([E]t=Ae−k1t+Be−k2t) by using the GraphPad Prism 6 software. For comparison, summarized in Table 4 are the biological half-lives (in mice/rats) of the native BChE (purified from human plasma) and recombinant BChE, BChE mutant, and CocH3-Fc(M3) produced using various methods. The biological half-life of Fc(M3)-(PAPAP)2—CocH3 is ˜105±7 hr, which is ˜6.6-fold longer than the biological half-life of recombinant BChE or mutant produced using the same method, ˜39-fold longer than the half-life of BChE produced in transgenic goat, and ˜525-fold longer than the BChE mutant produced in transgenic plant. Even compared to native human BChE, Fc(M3)-(PAPAP)2-CocH3 protein produced in CHO cells has a ˜2.4-fold prolonged biological half-life. In a previously reported study [31], it was demonstrated that a novel CocH form, i.e. a C-terminal Fc-fused CocH3, known as CocH3-Fc(M3), had a biological half-life of ˜107±6 hr in rats. But CocH3-Fc(M3) was expressed at a yield of ˜2.1 mg/L in CHO cells under the similar conditions. The Fc(M3)-(PAPAP)2—CocH3 reported in the current study has a similarly long half-life (105±7 hr vs 107±6 hr) compared to CocH3-Fc(M3) but with a significantly improved protein expression yield.









TABLE 4







Summary of biological half-life of BChE or


mutants in mice or rats











In vivo



Protein form
half-life (h)







BChE purified from human plasma
 43a



BChE produced in transgenic goat
 2.7b



BChE mutant produced in transgenic plant
 0.2c



BChE mutant produced in CHO cells
 7.3d



CocH3-Fc(M3) produced in CHO cells
107e



(SEQ ID NO: 13-SEQ ID NO: 16)




Fc(M3)-(PAPAP)2-CocH3 produced in CHO cells
105



(SEQ ID NO: 16-SEQ ID NO: 2-SEQ ID NO: 13)








aBiological half-life of enzyme was reported in ref. [26].





bBiological half-life of enzyme was reported in ref. [21].





cBiological half-life of enzyme was reported in ref. [19].





dData from ref. [24].





eData from ref. [31].







A previously reported study [31] demonstrated that a single injection of CocH-Fc(M3) was able to accelerate cocaine metabolism in rats after 20 days and, thus, block cocaine-induced physiological and toxic effects for a long period [31]. The CocH3-Fc(M3) protein was expected to allow dosing once every 2-4 wk, or longer, for treating cocaine addiction in humans. Given the facts that Fc(M3)-(PAPAP)2-CocH3 has the similarly long biological half-life in rats and same catalytic activity against cocaine, it is reasonable to expect that Fc(M3)-(PAPAP)2-CocH3 may also be able to provide the similar efficacy and duration for the cocaine addiction treatment.


It has been a significant challenge to efficiently express BChE polypeptides with both a long biological half-life comparable to the native BChE purified from human plasma and a high yield of protein expression. In this study, it has been demonstrated that an exemplary polypeptides including a BChE polypeptide molecule have not only a long biological half-life, but also an improved yield of protein expression compared to CocH3 (e.g., ˜105±7 hr in rats and ˜21-fold improved yield for Fc(M3)-(PAPAP)2-CocH3).


In a further example of the present invention:


BChE or BChE(574) refers to the wild-type human butyrylcholinesterase (full-length protein, with 574 amino acids) (SEQ ID NO:10). BChE(xxx) refers to a trucated fragment (with only the first xxx amino acids) of human butyrylcholinesterase (SEQ ID NO:10).


BChE-Fc refers to a fusion protein in which the C-terminus of human BChE (SEQ ID No: 10) is fused to the N-terminus of the Fc portion of human IgG-1 (SEQ ID NO: 8) or (SEQ ID NO 10-SEQ ID NO: 8). BChE(xxx)-Fc refers to a fusion protein in which the C-terminus of BChE(xxx) fragment fused to the N-terminus of the Fc portion of human IgG-1. CocH is a BChE polypeptide with specific mutations. CocH-LAF generally represents a cocaine hydrolase (CocH) in a long-acting form (LAF).









CocH-LAF1 (in which ″1″ means the first version)


refers to the


(SEQ ID NO: 13-SEQ ID NO: 16)


A199S/F227A/S287G/A328W/Y332G/A530V/D671E/L673M


mutant of BChE(529)-Fc.





CocH-LAF4 refers to the


(SEQ ID NO: 13-SEQ ID NO: 17)


A199S/F227A/S287G/A328W/Y332G/A530V/M567Y/D671E/


L673M mutant of the BChE(529)-Fc protein.





CocH-LAF6 refers to the


((SEQ ID NO: 13-SEQ ID NO: 18)


A199S/F227A/S287G/A328W/Y332G/A530V/M567Y/S569T/


T571E/D671E/L673M mutant of BChE(529)-Fc.





CocH-LAF7 refers to the


(SEQ ID NO: 14-SEQ ID NO: 18)


A199S/F227A/P285A/S287G/A328W/Y332G/A530V/M567Y/


S569T/T571E/D671E/L673M mutant of BChE(529)-Fc.





CocH-LAF8 refers to the


(SEQ ID NO: 15-SEQ ID NO: 18)


A199S/F227A/P285Q/S287G/A328W/Y332G/A530V/M567Y/


S569T/T571E/D671E/L673M mutant of BChE(529)-Fc.













TABLE 5







Transient expression levels of BChE-Fc and


BChE(529)-Fc










Expression



Protein
level (mg/L)
Ratio





BChE-Fc (SEQ ID NO: 11-SEQ
0.55
1


ID NO: 8)







BChE(529)-Fc (SEQ ID NO:
3.78
6.9


11-SEQ ID NO: 8)









According to the data in Table 5, BChE(529)-Fc can be expressed with a significantly improved yield (about ˜7 fold), compared BChE-Fc. Both the Fc fusion and BChE fragmentation did not significantly change the catalytic activity of BChE.


In light of the production data in Table 5, a further designed mutants of BChE(529)-Fc with an improved binding affinity with neonatal Fc receptor (FcRn) at pH 6 in order to further prolong the biological half-life (t1/2) in addition to the protein expression yield (see Table S2).









TABLE 6







Binding affinity of BChE(529)-Fc and its mutants with human FcRn


and their biological half-life (t1/2), along with the protein


expression levels in stably transfected CHO cells.











Kd (nM) with
t1/2 (hours)
Expression


Protein
FcRn at pH 6
in rats
levela





BChE(529)-Fc
2500 to 4000
 86 ± 6
>100 mg/L


(SEQ ID NO: 11-SEQ ID NO: 8)








CocH3-LAF1
 992
107 ± 6
  >1 g/L


(SEQ ID NO: 13-SEQ ID NO: 16)








CocH3-LAF4
 327
195 ± 10
  >1 g/L


(SEQ ID NO: 13-SEQ ID NO: 17)








CocH3-LAF6
  43
206 ± 7
>200 mg/L


(SEQ ID NO: 13-SEQ ID NO: 18)








CocHl-LAF7
  43
206 ± 7
  >1 g/L


(SEQ ID NO: 14-SEQ ID NO: 18)








CocH2-LAF8
  43
206 ± 7
>200 mg/L


(SEQ ID NO: 15-SEQ ID NO: 18)






aThe protein expression level is also affected by the culture conditions. Listed here is the lower end of the protein expression level.







To illustrate the approach to the rational design and discovery of BChE(529)-Fc mutants with improved binding affinity with FcRn and prolonged biological half-lives, depicted in FIG. 5 is the in vitro data for the binding affinity of CocH-LAF6.


CocH3 represents the A199S/F227A/S287G/A328W/Y332G mutant of human butyrylcholinesterase (BChE). The full-length BChE or CocH3 has 574 amino-acid residues. CocH3(xxx) refers to the fragment (with only the first xxx amino acids) of the A199S/F227A/S287G/A328W/Y332G mutant of human butyrylcholinesterase.









TABLE 7







Transient expression levels of fusion proteins









Expression 


Enzyme
level (mg/L)





CocH3(574) (SEQ ID NO: 12)
 0.51





Fc(M3)-CocH3(529)
 1.05


(SEQ ID NO: 16-SEQ ID NO: 13)






Fc(M3)-G3S-CocH3(529)
 3.10


(SEQ ID NO: 16-SEQ ID NO: 6-SEQ



ID NO: 13)






Fc(M3)-G4S-CocH3(529)
 3.69


(SEQ ID NO: 16-SEQ ID NO: 36-



SEQ ID NO: 13)






Fc(M3)-G6S-CocH3(529)
 5.14


(SEQ ID NO: 16-SEQ ID NO: 37-



SEQ ID NO: 13)






Fc(M3)-EAAAK-CocH3(529)
 4.76


(SEQ ID NO: 16-SEQ ID NO: 19-



SEQ ID NO: 13)






Fc(M3)-PAPAP-CocH3(529)
 5.15


(SEQ ID NO: 16-SEQ ID NO: 4-SEQ



ID NO: 13)






CocH3(529)-(G3S)2-Fc(M3)
 7.86


(SEQ ID NO: 13-SEQ ID NO: 6-SEQ



ID NO: 6-SEQ ID NO: 16)






Fc(M3)-(PAPAP)2-CocH3(529)
10.87


(SEQ ID NO: 16-SEQ ID NO: 2-SEQ



ID NO: 13)






CocH3(574)-Fc(M3)
 4.11


(SEQ ID NO: 12-SEQ ID NO: 16)






CocH3(574)-G6S-Fc(M3)
 3.96


(SEQ ID NO: 12-SEQ ID NO: 37-



SEQ ID NO: 16)






CocH3(529)-Fc(M3)
 5.75


(SEQ ID NO: 13-SEQ ID NO: 16)






CocH3(529)-G6S-Fc(M3)
 6.39


(SEQ ID NO: 13-SEQ ID NO: 37-



SEQ ID NO: 16)






CocH3(529)-Fc(M6)
 3.89


(SEQ ID NO: 13-SEQ ID NO: 18)









It will be understood that various details of the presently disclosed subject matter can be changed without departing from the scope of the subject matter disclosed herein. Furthermore, the foregoing description is for the purpose of illustration only, and not for the purpose of limitation.


All publications, patents, and patent applications mentioned in this specification are herein incorporated by reference to the same extent as if each individual publication, patent, or patent application was specifically and individually indicated to be incorporated by reference, including the references set forth in the following list:












SEQUENCE LISTING















SEQ ID NO: 1 Nucleotide Encoding SEQ ID NO: 2


CCGGCGCCGGCGCCGCCGGCGCCGGCGCCG





SEQ ID NO: 2


PAPAPPAPAP





SEQ ID NO: 3 Nucleotide encoding SEQ ID NO: 4


CCGGCGCCGGCGCCG





SEQ ID NO: 4


PAPAP





SEQ ID NO: 5 Nucleotide encoding SEQ ID NO: 6


GGCGGCGGCAGCGGCGGCGGCAGC





SEQ ID NO: 6


GGGSGGGS





SEQ ID NO: 7-Nucleotide encoding Wild type Fc polypeptide


GCA GAG CCT AAG TCC TGC GAC AAA ACT CAC ACA TGC CCA CCG TGC CCA


GCA CCT GAA CTC CTG GGG GGA CCG TCA GTC TTC CTC TTC CCC CCA AAA CCC


AAG GAC ACC CTC ATG ATC TCC CGG ACC CCT GAG GTC ACA TGC GTG GTG GTG


GAC GTG AGC CAC GAA GAC CCT GAG GTC AAG TTC AAC TGG TAC GTG GAC


GGC GTG GAG GTG CAT AAT GCC AAG ACA AAG CCG CGG GAG GAG CAG TAC


AAC AGC ACG TAC CGT GTG GTC AGC GTC CTC ACC GTC CTG CAC CAG GAC TGG


CTG AAT GGC AAG GAG TAC AAG TGC AAG GTC TCC AAC AAA GCC CTC CCA


GCC CCC ATC GAG AAA ACC ATC TCC AAA GCC AAA GGG CAG CCC CGA GAA


CCA CAG GTG TAC ACC CTG CCC CCA TCC CGG GAC GAG CTG ACC AAG AAC


CAG GTC AGC CTG ACC TGC CTG GTC AAA GGC TTC TAT CCC AGC GAC ATC GCC


GTG GAG TGG GAG AGC AAT GGG CAG CCG GAG AAC AAC TAC AAG ACC ACG


CCT CCC GTG CTG GAC TCC GAC GGC TCC TTC TTC CTC TAC AGC AAG CTC ACC


GTG GAC AAG AGC AGG TGG CAG CAG GGG AAC GTC TTC TCA TGC TCC GTG


ATG CAC GAG GCT CTG CAC AAC CAC TAC ACG CAG AAG AGC CTC TCC CTG TCT


CCG GGT AAA





SEQ ID NO: 8-Wild type Fc polypeptide


AEPKSCDKTH TCPPCPAPEL LGGPSVFLFP PKPKDTLMIS RTPEVTCVVV


DVSHEDPEVK FNWYVDGVEV HNAKTKPREE QYNSTYRVVS VLTVLHQDWL


NGKEYKCKVS NKALPAPIEK TISKAKGQPR EPQVYTLPPS RDELTKNQVS


LTCLVKGFYP SDIAVEWESN GQPENNYKTT PPVLDSDGSF FLYSKLTVDK


SRWQQGNVFS CSVMHEALHN HYTQKSLSLS PGK





SEQ ID NO: 9-Nucleotide encoding Wild type BChE


GAA GAT GAC ATC ATA ATT GCA ACA AAG AAT GGA AAA GTC AGA GGG ATG


AAC TTG ACA GTT TTT GGT GGC ACG GTA ACA GCC TTT CTT GGA ATT CCC TAT


GCA CAG CCA CCT CTT GGT AGA CTT CGA TTC AAA AAG CCA CAG TCT CTG ACC


AAG TGG TCT GAT ATT TGG AAT GCC ACA AAA TAT GCA AAT TCT TGC TGT CAG


AAC ATA GAT CAA AGT TTT CCA GGC TTC CAT GGA TCA GAG ATG TGG AAC CCA


AAC ACT GAC CTC AGT GAA GAC TGT TTA TAT CTA AAT GTA TGG ATT CCA GCA


CCT AAA CCA AAA AAT GCC ACT GTA TTG ATA TGG ATT TAT GGT GGT GGT TTT


CAA ACT GGA ACA TCA TCT TTA CAT GTT TAT GAT GGC AAG TTT CTG GCT CGG


GTT GAA AGA GTT ATT GTA GTG TCA ATG AAC TAT AGG GTG GGT GCC CTA GGA


TTC TTA GCT TTG CCA GGA AAT CCT GAG GCT CCA GGG AAC ATG GGT TTA TTT


GAT CAA CAG TTG GCT CTT CAG TGG GTT CAA AAA AAT ATA GCA GCC TTT GGT


GGA AAT CCT AAA AGT GTA ACT CTC TTT GGA GAA AGT GCA GGA GCA GCT TCA


GTT AGC CTG CAT TTG CTT TCT CCT GGA AGC CAT TCA TTG TTC ACC AGA GCC


ATT CTG CAA AGT GGT TCC TTT AAT GCT CCT TGG GCG GTA ACA TCT CTT TAT


GAA GCT AGG


AAC AGA ACG TTG AAC TTA GCT AAA TTG ACT GGT TGC TCT AGA GAG AAT GAG


ACT GAA ATA ATC AAG TGT CTT AGA AAT AAA GAT CCC CAA GAA ATT CTT CTG


AAT GAA GCA TTT GTT GTC CCC TAT GGG ACT CCT TTG TCA GTA AAC TTT GGT


CCG ACC GTG GAT GGT GAT TTT CTC ACT GAC ATG CCA GAC ATA TTA CTT GAA


CTT GGA CAA TTT AAA AAA ACC CAG ATT TTG GTG GGT GTT AAT AAA GAT GAA


GGG ACA GCT TTT TTA GTC TAT GGT GCT CCT GGC TTC AGC AAA GAT AAC AAT


AGT ATC ATA ACT AGA AAA GAA TTT CAG GAA GGT TTA AAA ATA TTT TTT CCA


GGA GTG AGT GAG TTT GGA AAG GAA TCC ATC CTT TTT CAT TAC ACA GAC TGG


GTA GAT GAT CAG AGA CCT GAA AAC TAC CGT GAG GCC TTG GGT GAT GTT GTT


GGG GAT TAT AAT TTC ATA TGC CCT GCC TTG GAG TTC ACC AAG AAG TTC TCA


GAA TGG GGA AAT AAT GCC TTT TTC TAC TAT TTT GAA CAC CGA TCC TCC AAA


CTT CCG TGG CCA GAA TGG ATG GGA GTG ATG CAT GGC TAT GAA ATT GAA TTT


GTC TTT GGT TTA CCT CTG GAA AGA AGA GAT AAT TAC ACA AAA GCC GAG


GAA ATT TTG AGT AGA TCC ATA GTG AAA CGG TGG GCA AAT TTT GCA AAA TAT


GGG AAT CCA


AAT GAG ACT CAG AAC AAT AGC ACA AGC TGG CCT GTC TTC AAA AGC ACT


GAA CAA AAA TAT CTA ACC TTG AAT ACA GAG TCA ACA AGA ATA ATG ACG


AAA CTA CGT GCT CAA CAA TGT CGA TTC TGG ACA TCA TTT TTT CCA AAA GTC


TTG GAA ATG ACA GGA AAT ATT GAT GAA GCA GAA TGG GAG TGG AAA GCA


GGA TTC CAT CGC TGG AAC AAT TAC ATG ATG GAC TGG AAA AAT CAA TTT AAC


GAT TAC ACT AGC AAG AAA GAA AGT TGT GTG GGT CTC





SEQ ID NO: 10-Wild type BChE Polypeptide


EDDIIIATKNGKVRGMNLTVFGGTVTAFLGIPYAQPPLGRLRFKKPQSLTKWSDIWNATK


YANSCCQNIDQSFPGFHGSEMWNPNTDLSEDCLYLNVWIPAPKPKNATVLIWIYGGGFQ


TGTSSLHVYDGKFLARVERVIVVSMNYRVGALGFLALPGNPEAPGNMGLFDQQLALQ


WVQKNIAAFGGNPKSVTLFGESAGAASVSLHLLSPGSHSLFTRAILQSGSFNAPWAVTSL


YEARNRTLNLAKLTGCSRENETEIIKCLRNKDPQEILLNEAFVVPYGTPLSVNFGPTVDG


DFLTDMPDILLELGQFKKTQILVGVNKDEGTAFLVYGAPGFSKDNNSIITRKEFQEGLKIF


FPGVSEFGKESILFHYTDWVDDQRPENYREALGDVVGDYNFICPALEFTKKFSEWGNNA


FFYYFERRSSKLPWPEWMGVMHGYEIEFVFGLPLERRDNYTKAEEILSRSIVKRWANFA


KYGNPNETQNNSTSWPVFKSTEQKYLTLNTESTRIMTKLRAQQCRFWTSFFPKVLEMTG


NIDEAEWEWKAGFHRWNNYMMDWKNQFNDYTSKKESCVGL





SEQ ID NO: 11-Truncated (only the first 529 amino acids of wild type BChE)


BchE (529)


EDDIIIATKNGKVRGMNLTVFGGTVTAFLGIPYAQPPLGRLRFKKPQSLTKWSDIWNATK


YANSCCQNIDQSFPGFHGSEMWNPNTDLSEDCLYLNVWIPAPKPKNATVLIWIYGGGFQ


TGTSSLHVYDGKFLARVERVIVVSMNYRVGALGFLALPGNPEAPGNMGLFDQQLALQ


WVQKNIAAFGGNPKSVTLFGESAGAASVSLHLLSPGSHSLFTRAILQSGSFNAPWAVTSL


YEARNRTLNLAKLTGCSRENETEIIKCLRNKDPQEILLNEAFVVPYGTPLSVNFGPTVDG


DFLTDMPDILLELGQFKKTQILVGVNKDEGTAFLVYGAPGFSKDNNSIITRKEFQEGLKIF


FPGVSEFGKESILFHYTDWVDDQRPENYREALGDVVGDYNFICPALEFTKKFSEWGNNA


FFYYFERRSSKLPWPEWMGVMHGYEIEFVFGLPLERRDNYTKAEEILSRSIVKRWANFA


KYGNPNETQNNSTSWPVFKSTEQKYLTLNTESTRIMTKLRAQQCRFWTSFFPKV





SEQ ID NO: 12-CoCH3 Full length (574)


EDDIIIATKNGKVRGMNLTVFGGTVTAFLGIPYAQPPLGRLRFKKPQSLTKWSDIWNATK


YANSCCQNIDQSFPGFHGSEMWNPNTDLSEDCLYLNVWIPAPKPKNATVLIWIYGGGFQ


TGTSSLHVYDGKFLARVERVIVVSMNYRVGALGFLALPGNPEAPGNMGLFDQQLALQ


WVQKNIAAFGGNPKSVTLFGESSGAASVSLHLLSPGSHSLFTRAILQSGSANAPWAVTSL


YEARNRTLNLAKLTGCSRENETEIIKCLRNKDPQEILLNEAFVVPYGTPLGVNFGPTVDG


DFLTDMPDILLELGQFKKTQILVGVNKDEGTWFLVGGAPGFSKDNNSIITRKEFQEGLKI


FFPGVSEFGKESILFHYTDWVDDQRPENYREALGDVVGDYNFICPALEFTKKFSEWGNN


AFFYYFEHRSSKLPWPEWMGVMHGYEIEFVFGLPLERRDNYTKAEEILSRSIVKRWANF


AKYGNPNETQNNSTSWPVFKSTEQKYLTLNTESTRIMTKLRAQQCRFWTSFFPKVLEMT


GNIDEAEWEWKAGFHRWNNYMMDWKNQFNDYTSKKESCVGL





SEQ ID NO: 13-Truncated CoCH3 (only the first 529 amino acids of Full length


CoCH3)(529)


EDDIIIATKNGKVRGMNLTVFGGTVTAFLGIPYAQPPLGRLRFKKPQSLTKWSDIWNATK


YANSCCQNIDQSFPGFHGSEMWNPNTDLSEDCLYLNVWIPAPKPKNATVLIWIYGGGFQ


TGTSSLHVYDGKFLARVERVIVVSMNYRVGALGFLALPGNPEAPGNMGLFDQQLALQ


WVQKNIAAFGGNPKSVTLFGESSGAASVSLHLLSPGSHSLFTRAILQSGSANAPWAVTSL


YEARNRTLNLAKLTGCSRENETEIIKCLRNKDPQEILLNEAFVVPYGTPLGVNFGPTVDG


DFLTDMPDILLELGQFKKTQILVGVNKDEGTWFLVGGAPGFSKDNNSIITRKEFQEGLKI


FFPGVSEFGKESILFHYTDWVDDQRPENYREALGDVVGDYNFICPALEFTKKFSEWGNN


AFFYYFEHRSSKLPWPEWMGVMHGYEIEFVFGLPLERRDNYTKAEEILSRSIVKRWANF


AKYGNPNETQNNSTSWPVFKSTEQKYLTLNTESTRIMTKLRAQQCRFWTSFFPKV





SEQ ID NO: 14-CoCH1


EDDIIIATKNGKVRGMNLTVFGGTVTAFLGIPYAQPPLGRLRFKKPQSLTKWSDIWNATK


YANSCCQNIDQSFPGFHGSEMWNPNTDLSEDCLYLNVWIPAPKPKNATVLIWIYGGGFQ


TGTSSLHVYDGKFLARVERVIVVSMNYRVGALGFLALPGNPEAPGNMGLFDQQLALQ


WVQKNIAAFGGNPKSVTLFGESSGAASVSLHLLSPGSHSLFTRAILQSGSANAPWAVTSL


YEARNRTLNLAKLTGCSRENETEIIKCLRNKDPQEILLNEAFVVPYGTALGVNFGPTVDG


DFLTDMPDILLELGQFKKTQILVGVNKDEGTWFLVGGAPGFSKDNNSIITRKEFQEGLKI


FFPGVSEFGKESILFHYTDWVDDQRPENYREALGDVVGDYNFICPALEFTKKFSEWGNN


AFFYYFEHRSSKLPWPEWMGVMHGYEIEFVFGLPLERRDNYTKAEEILSRSIVKRWANF


AKYGNPNETQNNSTSWPVFKSTEQKYLTLNTESTRIMTKLRAQQCRFWTSFFPKV





SEQ ID NO: 15-CoCH2


EDDIIIATKNGKVRGMNLTVFGGTVTAFLGIPYAQPPLGRLRFKKPQSLTKWSDIWNATK


YANSCCQNIDQSFPGFHGSEMWNPNTDLSEDCLYLNVWIPAPKPKNATVLIWIYGGGFQ


TGTSSLHVYDGKFLARVERVIVVSMNYRVGALGFLALPGNPEAPGNMGLFDQQLALQ


WVQKNIAAFGGNPKSVTLFGESSGAASVSLHLLSPGSHSLFTRAILQSGSANAPWAVTSL


YEARNRTLNLAKLTGCSRENETEIIKCLRNKDPQEILLNEAFVVPYGTQLGVNFGPTVDG


DFLTDMPDILLELGQFKKTQILVGVNKDEGTWFLVGGAPGFSKDNNSIITRKEFQEGLKI


FFPGVSEFGKESILFHYTDWVDDQRPENYREALGDVVGDYNFICPALEFTKKFSEWGNN


AFFYYFEHRSSKLPWPEWMGVMHGYEIEFVFGLPLERRDNYTKAEEILSRSIVKRWANF


AKYGNPNETQNNSTSWPVFKSTEQKYLTLNTESTRIMTKLRAQQCRFWTSFFPKV





SEQ ID NO: 16-Fc(M3)


VEPKSCDKTH TCPPCPAPEL LGGPSVFLFP PKPKDTLMIS RTPEVTCVVV


DVSHEDPEVK FNWYVDGVEV HNAKTKPREE QYNSTYRVVS VLTVLHQDWL


NGKEYKCKVS NKALPAPIEK TISKAKGQPR EPQVYTLPPS REEMTKNQVS


LTCLVKGFYP SDIAVEWESN GQPENNYKTT PPVLDSDGSF FLYSKLTVDK


SRWQQGNVFS CSVMHEALHN HYTQKSLSLS PGK





SEQ ID NO: 17-Fc(M4)


VEPKSCDKTH TCPPCPAPEL LGGPSVFLFP PKPKDTYIS RTPEVTCVVV


DVSHEDPEVK FNWYVDGVEV HNAKTKPREE QYNSTYRVVS VLTVLHQDWL


NGKEYKCKVS NKALPAPIEK TISKAKGQPR EPQVYTLPPS REEMTKNQVS


LTCLVKGFYP SDIAVEWESN GQPENNYKTT PPVLDSDGSF FLYSKLTVDK


SRWQQGNVFS CSVMHEALHN HYTQKSLSLS PGK





SEQ ID NO: 18-Fc(M6)


VEPKSCDKTH TCPPCPAPEL LGGPSVFLFP PKPKDTYIT REPEVTCVVV


DVSHEDPEVK FNWYVDGVEV HNAKTKPREE QYNSTYRVVS VLTVLHQDWL


NGKEYKCKVS NKALPAPIEK TISKAKGQPR EPQVYTLPPS REEMTKNQVS


LTCLVKGFYP SDIAVEWESN GQPENNYKTT PPVLDSDGSF FLYSKLTVDK


SRWQQGNVFS CSVMHEALHN HYTQKSLSLS PGK





SEQ ID NO: 19-EAAAK Polypeptide


EAAAK





SEQ ID NO: 20-EAAAK-Forward Primer


G TCT CCG GGT AAA GAG GCT GCC GCC AAG GAA GAT GAC ATC A





SEQ ID NO: 21-EAAAK-Reverse Primer


CTT GGC GGC AGC CTC TTT ACC CGG AGA CAG GGA GAG





SEQ ID NO: 22-PAPAP-Forward Primer


G TCT CCG GGT AAA CCT GCT CCA GCC CCG GAA GAT GAC ATC





SEQ ID NO: 23-PAPAP-Reverse Primer


CGG GGC TGG AGC AGG TTT ACC CGG AGA CAG GGA GAG





SEQ ID NO: 24-(G3S)2-Forward Primer


G TCT CCG GGT AAA GGT GGA GGT TCC GGT GGA GGT TCC GAA GAT GAC ATC A





SEQ ID NO: 25-(G3S)2-Reverse Primer


GGA ACC TCC ACC GGA ACC TCC ACC TTT ACC CGG AGA CAG GGA GAG





SEQ ID NO: 26-(PAPAP)2-Forward Primer


G TCT CCG GGT AAA CCT GCT CCA GCC CCG CCT GCT CCA GCC CCG GAA GAT


GAC ATC





SEQ ID NO: 27-(PAPAP)2-Reverse Primer


CGG GGC TGG AGC AGG CGG GGC TGG AGC AGG TTT ACC CGG AGA CAG GGA


GAG





SEQ ID NO: 28-Fc(M8)


VEPKSCDKTH TCPPCPAPEL LGGPSVFLFP PKPKDTLMIS RTPEVTCVVV


DVSHEDPQVK FNWYVDGVQV HNAKTKPREQ QYNSTYRVVS VLTVLHQNWL


DGKEYKCKVS NKALPAPIEK TISKAKGQPR EPQVYTLPPS REEMTKNQVS


LTCLVKGFYP SDIAVEWESN GQPENNYKTT PPVLDSDGSF FLYSKLTVDK


SRWQQGNVFS CSVMHEALHN HYTQKSLSLS PGK





SEQ ID NO: 29-Fc(M5)


QEPKSSDKTH TSPPSPAPEL LGGSSVFLFP PKPKDTLMIS RTPEVTCVVV


DVSHEDPEVK FNWYVDGVEV HNAKTKPREE QYNSTYRVVS VLTVLHQDWL


NGKEYKCKVS NKALPAPIEK TISKAKGQPR EPQVYTLPPS RDELTKNQVS


LTCLVKGFYP SDIAVEWESN GQPENNYKTT PPVLDSDGSF FLYSKLTVDK


SRWQQGNVFS CSVMHEALHN HYTQKSLSLS PGK





SEQ ID NO: 30-Fc(M4′)


VEPKSCDKTH TCPPCPAPEL LGGPSVFLFP PKPKDTLMIS REPEVTCVVV


DVSHEDPEVK FNWYVDGVEV HNAKTKPREE QYNSTYRVVS VLTVLHQDWL


NGKEYKCKVS NKALPAPIEK TISKAKGQPR EPQVYTLPPS REEMTKNQVS


LTCLVKGFYP SDIAVEWESN GQPENNYKTT PPVLDSDGSF FLYSKLTVDK


SRWQQGNVFS CSVMHEALHN HYTQKSLSLS PGK





SEQ ID NO: 31-Fc(M5′)


VEPKSCDKTH TCPPCPAPEL LGGPSVFLFP PKPKDTLYIT RTPEVTCVVV


DVSHEDPEVK FNWYVDGVEV HNAKTKPREE QYNSTYRVVS VLTVLHQDWL


NGKEYKCKVS NKALPAPIEK TISKAKGQPR EPQVYTLPPS REEMTKNQVS


LTCLVKGFYP SDIAVEWESN GQPENNYKTT PPVLDSDGSF FLYSKLTVDK


SRWQQGNVFS CSVMHEALHN HYTQKSLSLS PGK





SEQ ID NO: 32-Fc(M6′)


QEPKSSDKTH TSPPSPAPEL LGGSSVFLFP PKPKDTLYIS RTPEVTCVVV


DVSHEDPEVK FNWYVDGVEV HNAKTKPREE QYNSTYRVVS VLTVLHQDWL


NGKEYKCKVS NKALPAPIEK TISKAKGQPR EPQVYTLPPS RDELTKNQVS


LTCLVKGFYP SDIAVEWESN GQPENNYKTT PPVLDSDGSF FLYSKLTVDK


SRWQQGNVFS CSVMHEALHN HYTQKSLSLS PGK





SEQ ID NO: 33-Fc(M7)


QEPKSSDKTH TSPPSPAPEL LGGSSVFLFP PKPKDTLYIT RTPEVTCVVV


DVSHEDPEVK FNWYVDGVEV HNAKTKPREE QYNSTYRVVS VLTVLHQDWL


NGKEYKCKVS NKALPAPIEK TISKAKGQPR EPQVYTLPPS RDELTKNQVS


LTCLVKGFYP SDIAVEWESN GQPENNYKTT PPVLDSDGSF FLYSKLTVDK


SRWQQGNVFS CSVMHEALHN HYTQKSLSLS PGK





SEQ ID NO: 34-Fc(M8′)


QEPKSSDKTH TSPPSPAPEL LGGSSVFLFP PKPKDTLYIT REPEVTCVVV


DVSHEDPEVK FNWYVDGVEV HNAKTKPREE QYNSTYRVVS VLTVLHQDWL


NGKEYKCKVS NKALPAPIEK TISKAKGQPR EPQVYTLPPS RDELTKNQVS


LTCLVKGFYP SDIAVEWESN GQPENNYKTT PPVLDSDGSF FLYSKLTVDK


SRWQQGNVFS CSVMHEALHN HYTQKSLSLS PGK





SEQ ID NO: 35-wild type Fc-Truncated wild type BChE-fusion polypeptide


AEPKSCDKTH TCPPCPAPEL LGGPSVFLFP PKPKDTLMIS


RTPEVTCVVVDVSHEDPEVK FNWYVDGVEV HNAKTKPREE QYNSTYRVVS


VLTVLHQDWL NGKEYKCKVS NKALPAPIEK TISKAKGQPR EPQVYTLPPS


RDELTKNQVS LTCLVKGFYP SDIAVEWESN GQPENNYKTT PPVLDSDGSF


FLYSKLTVDK SRWQQGNVFS CSVMHEALHN HYTQKSLSLS


PGKEDDIIIATKNGKVRGMNLTVFGGTVTAFLGIPYAQPPLGRLRFKKPQSLTKWSDIWN


ATKYANSCCQNIDQSFPGFHGSEMWNPNTDLSEDCLYLNVWIPAPKPKNATVLIWIYGG


GFQTGTSSLHVYDGKFLARVERVIVVSMNYRVGALGFLALPGNPEAPGNMGLFDQQLA


LQWVQKNIAAFGGNPKSVTLFGESAGAASVSLHLLSPGSHSLFTRAILQSGSFNAPWAV


TSLYEARNRTLNLAKLTGCSRENETEIIKCLRNKDPQEILLNEAFVVPYGTPLSVNFGPTV


DGDFLTDMPDILLELGQFKKTQILVGVNKDEGTAFLVYGAPGFSKDNNSIITRKEFQEGL


KIFFPGVSEFGKESILFHYTDWVDDQRPENYREALGDVVGDYNFICPALEFTKKFSEWG


NNAFFYYFEHRSSKLPWPEWMGVMHGYEIEFVFGLPLERRDNYTKAEEILSRSIVKRWA


NFAKYGNPNETQNNSTSWPVFKSTEQKYLTLNTESTRIMTKLRAQQCRFWTSFFPKV





SEQ ID NO: 36-G4S


GGGGS GGGGS





SEQ ID NO: 37-G6S


GGGGGGS GGGGGGS









REFERENCES



  • [1] O. Lockridge, Review of human butyrylcholinesterase structure, function, genetic variants, history of use in the clinic, and potential therapeutic uses, Pharmacol Ther, 148 (2015) 34-46.

  • [2] D. Cerasoli, E. Griffiths, B. Doctor, A. Saxena, J. Fedorko, N. Greig, Q. Yu, Y. Huang, H. Wilgus, C. Karatzas, (07) In vitro and in vivo characterization of recombinant human butyrylcholinesterase (Protexia™) as a potential nerve agent bioscavenger, Chemico-biological interactions, 157 (2005) 362-365.

  • [3] D. E. Lenz, D. Yeung, J. R. Smith, R. E. Sweeney, L. A. Lumley, D. M. Cerasoli, Stoichiometric and catalytic scavengers as protection against nerve agent toxicity: a mini review, Toxicology, 233 (2007) 31-39.

  • [4] Y. Ashani, S. Shapira, D. Levy, A. D. Wolfe, B. P. Doctor, Butyrylcholinesterase and acetylcholinesterase prophylaxis against soman poisoning in mice, Biochem Pharmacol, 41 (1991) 37-41.

  • [5] R. Brandeis, L. Raveh, J. Grunwald, E. Cohen, Y. Ashani, Prevention of soman-induced cognitive deficits by pretreatment with human butyrylcholinesterase in rats, Pharmacology Biochemistry and Behavior, 46 (1993) 889-896.

  • [6] N. Allon, L. Raveh, E. Gilat, E. Cohen, J. Grunwald, Y. Ashani, Prophylaxis against soman inhalation toxicity in guinea pigs by pretreatment alone with human serum butyrylcholinesterase, Toxicol Sci, 43 (1998) 121-128.

  • [7] H. Sun, Y.-P. Pang, O. Lockridge, S. Brimijoin, Re-engineering Butyrylcholinesterase as a Cocaine Hydrolase, Mol Pharmacol, 62 (2002) 220-224.

  • [8] S. Brimijoin, Y. Gao, J. J. Anker, L. A. Gliddon, D. LaFleur, R. Shah, Q. Zhao, M. Singh, M. E. Carroll, A Cocaine Hydrolase Engineered from Human Butyrylcholinesterase Selectively Blocks Cocaine Toxicity and Reinstatement of Drug Seeking in Rats, Neuropsychopharmacology, 33 (2008) 2715-2725.

  • [9] F. Zheng, W. C. Yang, M. C. Ko, J. J. Liu, H. Cho, D. Q. Gao, M. Tong, H. H. Tai, J. H. Woods, C.-G. Zhan, Most Efficient Cocaine Hydrolase Designed by Virtual Screening of Transition States, J Am Chem Soc, 130 (2008) 12148-12155.

  • [10] L. Xue, M.-C. Ko, M. Tong, W. Yang, S. Hou, L. Fang, J. Liu, F. Zheng, J. H. Woods, H.-H. Tai, C.-G. Zhan, Design, preparation, and characterization of high-activity mutants of human butyrylcholinesterase specific for detoxification of cocaine, Mol Pharmacol, 79 (2011) 290-297.

  • [11] L. Fang, F. Zheng, C.-G. Zhan, A model of glycosylated human butyrylcholinesterase, Mol. BioSyst., 10 (2014) 348-354.

  • [12] Y. Pan, D. Gao, W. Yang, H. Cho, G. Yang, H.-H. Tai, C.-G. Zhan, Computational redesign of human butyrylcholinesterase for anticocaine medication, Proc Natl Acad Sci USA, 102 (2005) 16656-16661.

  • [13] O. Cohen-Barak, J. Wildeman, J. van de Wetering, J. Hettinga, P. Schuilenga-Hut, A. Gross, S. Clark, M. Bassan, Y. Gilgun-Sherki, B. Mendzelevski, O. Spiegelstein, Safety, Pharmacokinetics, and Pharmacodynamics of TV-1380, a Novel Mutated Butyrylcholinesterase Treatment for Cocaine Addiction, After Single and Multiple Intramuscular Injections in Healthy Subjects, J Clin Pharmacol, 55 (2015) 573-583.

  • [14] M. J. Shram, O. Cohen-Barak, B. Chakraborty, M. Bassan, K. A. Schoedel, H. Hallak, E. Eyal, S. Weiss, Y. Gilgun, E. M. Sellers, J. Faulknor, O. Spiegelstein, Assessment of Pharmacokinetic and Pharmacodynamic Interactions Between Albumin-Fused Mutated Butyrylcholinesterase and Intravenously Administered Cocaine in Recreational Cocaine Users, J Clin Psychopharmacol, 35 (2015) 396-405.

  • [15] F. Zheng, C.-G. Zhan, Are pharmacokinetic approaches feasible for treatment of cocaine addiction and overdose?, Future Med Chem, 4 (2012) 125-128.

  • [16] P. Masson, A. Steve, P.-t. Philippe, L. Oksana, Multidisciplinary approaches to cholinesterase functions. expression and refoldin of functional human butyrylcholinesterase in, E. coli.

  • [17] W.-L. Wei, J.-C. Qin, M.-J. Sun, High-level expression of human butyrylcholinesterase gene in Bombyx mori and biochemical-pharmacological characteristic study of its product, Biochemical pharmacology, 60 (2000) 121-126.

  • [18] X. Brazzolotto, M. Wandhammer, C. Ronco, M. Trovaslet, L. Jean, O. Lockridge, P. Y. Renard, F. Nachon, Human butyrylcholinesterase produced in insect cells: huprine-based affinity purification and crystal structure, FEBS Journal, 279 (2012) 2905-2916.

  • [19] G. Wang, T. Zhang, H. Huang, S. Hou, X. Chen, F. Zheng, C.-G. Zhan, Plant expression of cocaine hydrolase-Fc fusion protein for treatment of cocaine abuse, BMC biotechnology, 16 (2016) 72.

  • [20] B. C. Geyer, L. Kannan, I. Cherni, R. R. Woods, H. Soreq, T. S. Mor, Transgenic plants as a source for the bioscavenging enzyme, human butyrylcholinesterase, Plant biotechnology journal, 8 (2010) 873-886.

  • [21] Y.-J. Huang, Y. Huang, H. Baldassarre, B. Wang, A. Lazaris, M. Leduc, A. S. Bilodeau, A. Bellemare, M. Cote, P. Herskovits, Recombinant human butyrylcholinesterase from milk of transgenic animals to protect against organophosphate poisoning, Proc Natl Acad Sci USA, 104 (2007) 13603-13608.

  • [22] Y.-J. Huang, P. M. Lundy, A. Lazaris, Y. Huang, H. Baldassarre, B. Wang, C. Turcotte, M. Côté, A. Bellemare, A. S. Bilodeau, Substantially improved pharmacokinetics of recombinant human butyrylcholinesterase by fusion to human serum albumin, BMC biotechnology, 8 (2008) 50.

  • [23] F. Li, N. Vijayasankaran, A. Shen, R. Kiss, A. Amanullah, Cell culture processes for monoclonal antibody production, MAbs, Taylor & Francis, 2010, pp. 466-479.

  • [24] L. Xue, S. Hou, M. Tong, L. Fang, X. Chen, Z. Jin, H.-H. Tai, F. Zheng, C.-G. Zhan, Preparation and in vivo characterization of a cocaine hydrolase engineered from human butyrylcholinesterase for metabolizing cocaine, Biochem J, 453 (2013) 447-454.

  • [25] E. G. Duysen, C. F. Bartels, O. Lockridge, Wild-type and A328W mutant human butyrylcholinesterase tetramers expressed in Chinese hamster ovary cells have a 16-hour half-life in the circulation and protect mice from cocaine toxicity, Journal of Pharmacology and Experimental Therapeutics, 302 (2002) 751-758.

  • [26] 0. Lockridge, L. M. Schopfer, G. Winger, J. H. Woods, Large scale purification of butyrylcholinesterase from human plasma suitable for injection into monkeys; a potential new therapeutic for protection against cocaine and nerve agent toxicity, The journal of medical, chemical, biological, and radiological defense, 3 (2005) nihms5095.

  • [27] E. R. Lavallie, E. A. DiBlasio, S. Kovacic, K. L. Grant, P. F. Schendel, J. M. McCoy, A thioredoxin gene fusion expression system that circumvents inclusion body formation in the E. coli cytoplasm, Nature biotechnology, 11 (1993) 187-193.

  • [28] K.-M. Lo, Y. Sudo, J. Chen, Y. Li, Y. Lan, S.-M. Kong, L. Chen, Q. An, S. D. Gillies, High level expression and secretion of Fc-X fusion proteins in mammalian cells, Protein engineering, 11 (1998) 495-500.

  • [29] D. C. Roopenian, S. Akilesh, FcRn: the neonatal Fc receptor comes of age, Nature reviews immunology, 7 (2007) 715-725.

  • [30] W. Wang, J. Brady, K. Donato, M. Peshwa, K. Steger, Gram Scale Antibody Production Using CHO Cell Transient Gene Expression (TGE) via Flow Electroporation, J. Biomol. Screen, 20 (2015) 545-551.

  • [31] X. Chen, L. Xue, S. Hou, Z. Jin, T. Zhang, F. Zheng, C.-G. Zhan, Long-acting cocaine hydrolase for addiction therapy, Proc Natl Acad Sci USA, 113 (2016) 422-427.

  • [32] X. Chen, W. Cui, J. Deng, S. Hou, J. Zhang, X. Ding, X. Zheng, H. Wei, Z. Zhou, K. Kim, C.-G. Zhan, F. zheng, Development of Fc-fused Cocaine Hydrolase for Cocaine Addiction: Catalytic and Pharmacokinetic Properties, AAPS J, 20 (2018) 53. doi: 10.1208/s12248-12018-10214-12249.

  • [33] X. Chen, X. Huang, L. Geng, L. Xue, S. Hou, X. Zheng, S. Brimijoin, F. Zheng, C.-G. Zhan, Kinetic characterization of a cocaine hydrolase engineered from mouse butyrylcholinesterase, Biochemical Journal, 466 (2015) 243-251.


Claims
  • 1. A fusion polypeptide, comprising: an Fc polypeptide joined to an N- or C-terminal end of a butyrylcholinesterase (BChE) polypeptide, wherein (a) the Fc polypeptide is joined to the BChE polypeptide via a linker, the linker comprising a sequence selected from the sequences of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 19, and SEQ ID NO: 37;(b) the Fc polypeptide has the sequence of SEQ ID NO: 8, or a fragment thereof, wherein the Fc polypeptide or fragment thereof includes 3 to 8 amino acid substitutions at 3 to 8 of residues selected from 1, 6, 12, 15, 24, 38, 40, 42, 58, 69, 80, 98, 101, 142, and 144; and(c) the fusion polypeptide has an improved production yield and biological half-life compared to a wild-type BChE polypeptide which is not fused.
  • 2. The fusion polypeptide of claim 1, wherein the Fc polypeptide is a fragment wherein 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16 amino acids are removed from the N-terminus of SEQ ID NO: 8.
  • 3. The fusion polypeptide of claim 1, wherein the Fc polypeptide is selected from SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 28, SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 31, SEQ ID NO: 32, SEQ ID NO: 33, and SEQ ID NO: 34.
  • 4. The fusion polypeptide of claim 1, wherein the BChE polypeptide has the sequence of SEQ ID NO: 10 or a fragment thereof, wherein the BChE polypeptide or fragment thereof includes 3 to 8 amino acid substitutions at 3 to 8 of residues chosen from 199, 227, 285, 286, 287, 328, 332, and 441.
  • 5. The fusion polypeptide of claim 4, wherein the BChE polypeptide has a group of amino acid substitutions selected from A199S, F227A, F227S, F227Q, F227I, F227G, F227V, F227I, F227L, F227S, F227T, F227M, F227C, P285A, P285S, P285Q, P285I, P285G, P285M, P285N, P285E, S287G, A328W, Y332G, E441D, and combinations thereof.
  • 6. The fusion polypeptide of claim 5, wherein the BChE polypeptide is a fragment wherein from 1 to 116 amino acids are removed from the N-terminus of SEQ ID NO: 10.
  • 7. The fusion polypeptide of claim 5, wherein the BChE polypeptide is a fragment wherein from 1 to 432 amino acids are removed from the C-terminus of SEQ ID NO: 10.
  • 8. The fusion polypeptide of claim 7, wherein the BChE polypeptide has a group of amino acid substitutions selected from A199S, F227A, F227S, F227Q, F227I, F227G, F227V, F227I, F227L, F227S, F227T, F227M, F227C, P285A, P285S, P285Q, P285I, P285G, P285M, P285N, P285E, S287G, A328W, Y332G, E441D, and combinations thereof.
  • 9. The fusion polypeptide of claim 1, wherein the transient expression level of the polypeptide is at least 9 times higher than a reference BChE polypeptide that does not include the Fc polypeptide and linker.
  • 10. The fusion polypeptide of claim 1 of SEQ ID NO: 35.
  • 11. The fusion polypeptide of claim 1, wherein the BChE polypeptide is selected from: SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, and SEQ ID NO: 15.
  • 12. The fusion polypeptide of claim 11, wherein the Fc polypeptide is selected from SEQ ID NO: 16, SEQ ID NO: 17, and SEQ ID NO: 18.
  • 13. The fusion polypeptide of claim 1, wherein the transient expression level of the polypeptide is about 9 times higher than a reference BChE polypeptide that does not include the Fc polypeptide and linker.
RELATED APPLICATIONS

This application claims priority from U.S. Provisional Patent Application No. 62/950,765 filed on Dec. 19, 2019 the entire disclosure of which is incorporated herein by this reference.

GOVERNMENT INTEREST

This invention was made with government support under Grant Numbers DA041115, DA035552, DA032910, DA013930, and DA025100 awarded by the National Institutes of Health (NIH). The government has certain rights in the invention.

US Referenced Citations (1)
Number Name Date Kind
20190255155 Perlroth Aug 2019 A1
Non-Patent Literature Citations (6)
Entry
Cerasoli, et ao., In vitro and in vivo characterization of recombinant human butyrylcholinesterase (Protexia™) as a potential nerve agent bioscavenger, Chemico-biological interactions, 157 (2005) 362-365.
Ashani, et al., Butyrylcholinesterase and acetylcholinesterase prophylaxis against soman poisoning in mice, Biochem Pharmacol, 41 (1991) 37-41.
Brazzolotto, et al., Human butyrylcholinesterase produced in insect cells: huprine-based affinity purification and crystal structure, FEBS Journal, 279 (2012) 2905-2916.
Geyer, et al., Transgenic plants as a source for the bioscavenging enzyme, human butyrylcholinesterase, Plant biotechnology journal, 8 (2010) 873-886.
Huang, et al., Recombinant human butyrylcholinesterase from milk of transgenic animals to protect against organophosphate poisoning, Proc Natl Acad Sci U S A, 104 (2007) 13603-13608.
Nachon, et al., Engineering of a monomeric and low-glycosylated form of human butyrylcholinesterase, Eur. J. Biochem. 269, 630-637 (2002).
Related Publications (1)
Number Date Country
20210189359 A1 Jun 2021 US
Provisional Applications (1)
Number Date Country
62950765 Dec 2019 US