Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
ENGINEERED BOTULINUM NEUROTOXIN
Document Type and Number:
WIPO Patent Application WO/2016/154534
Kind Code:
A1
Abstract:
Disclosed herein is a botulinum neurotoxin (BoNT) polypeptide with a modified receptor binding domain (HC) having one or more amino acid mutations that modify the binding of the BoNT to the receptor. Specific mutations and combinations of mutations are also disclosed. Isolated modified HC, polypeptides comprising the modified HC, chimeric molecules, pharmaceutical compositions, and methods of making and using the same are also disclosed. Methods of identifying additional such modified receptor binding domains, are further disclosed.

Inventors:
DONG MIN (US)
PENG LISHENG (CN)
TAO LIANG (US)
Application Number:
PCT/US2016/024211
Publication Date:
September 29, 2016
Filing Date:
March 25, 2016
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
HARVARD COLLEGE (US)
International Classes:
C07K14/33; A61K38/48
Domestic Patent References:
WO2013180799A12013-12-05
WO1995032738A11995-12-07
WO1996033273A11996-10-24
WO1998007864A11998-02-26
WO1999017806A11999-04-15
WO2006001676A12006-01-05
WO2008059126A12008-05-22
WO2008090287A22008-07-31
WO2009130600A22009-10-29
WO2007144493A22007-12-21
Foreign References:
US8052979B22011-11-08
US20070020295A12007-01-25
US20100196445A12010-08-05
US5721215A1998-02-24
US6113915A2000-09-05
US5766605A1998-06-16
US5714468A1998-02-03
Other References:
A. RUMMEL ET AL: "Identification of the protein receptor binding site of botulinum neurotoxins B and G proves the double-receptor concept", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES, vol. 104, no. 1, 2 January 2007 (2007-01-02), pages 359 - 364, XP055069704, ISSN: 0027-8424, DOI: 10.1073/pnas.0609713104
DATABASE UniProt [online] 26 June 2013 (2013-06-26), "SubName: Full=Neurotoxin B8 {ECO:0000313|EMBL:AGJ83978.1};", XP002759279, retrieved from EBI accession no. UNIPROT:M9VUL2 Database accession no. M9VUL2
DATABASE Geneseq [online] 13 September 2012 (2012-09-13), "Modified Clostridial toxin construct BoNT/B-HA-17 beta.", XP002759280, retrieved from EBI accession no. GSP:AZY12935 Database accession no. AZY12935
RONNIE P-A. BERNTSSON ET AL: "Structure of dual receptor binding to botulinum neurotoxin B", NATURE COMMUNICATIONS, vol. 4, 28 June 2013 (2013-06-28), XP055283679, DOI: 10.1038/ncomms3058
ARNON, S. ET AL., JAMA, vol. 285, 2001, pages 1059
CHAI ET AL., NATURE, vol. 444, pages 1096
PENG ET AL., J CELL SCIENCE, 2012
HILL ET AL., JOURNAL OF BACTERIOLOGY, vol. 189, no. 3, 2007, pages 818 - 832
YANN HUMEAU ET AL.: "How Botulinum and Tetanus Neurotoxins Block Neurotransmitter Release", BIOCHIMIE, vol. 82, no. 5, 2000, pages 427 - 446
KATHRYN TURTON ET AL.: "Botulinum and Tetanus Neurotoxins: Structure, Function and Therapeutic Utility", TRENDS BIOCHEM. SCI., vol. 27, no. 11, 2002, pages 552 - 558, XP004390740, DOI: doi:10.1016/S0968-0004(02)02177-1
GIOVANNA LALLI ET AL.: "The Journey of Tetanus and Botulinum Neurotoxins in Neurons", TRENDS MICROBIOL., vol. 11, no. 9, 2003, pages 431 - 437
SAWYER: "Peptide Based Drug Design", 1995, ACS, pages: 378 - 422
"INSECT CELL CULTURE ENGINEERING", 1993, MARCEL DEKKER
"INSECT CELL CULTURES: FUNDAMENTAL AND APPLIED ASPECTS", 1996, KLUWER ACADEMIC PUBLISHERS
MAUREEN A. HARRISON; IAN F. RAE: "GENERAL TECHNIQUES OF CELL CULTURE", 1997, CAMBRIDGE UNIVERSITY PRESS
"CELL AND TISSUE CULTURE: LABORATORY PROCEDURES", 1998, JOHN WILEY AND SONS
R. IAN FRESHNEY: "CULTURE OF ANIMAL CELLS: A MANUAL OF BASIC TECHNIQUE", 2000, WILEY-LISS
"ANIMAL CELL CULTURE: A PRACTICAL APPROACH", 2000, OXFORD UNIVERSITY PRESS
"MOLECULAR CLONING A LABORATORY MANUAL", 2001
"BASIC CELL CULTURE: A PRACTICAL APPROACH", 2002, OXFORD PRESS
"CURRENT PROTOCOLS IN MOLECULAR BIOLOGY", 2004
PETER F. BONVENTRE; LLOYD L. KLEMPE, J. BACTERIOL, vol. 79, no. 1, 1960, pages 23
MICHAEAL L. DEKLEVA; BIBHUTI R. DASGUPTA, BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICAITONS, vol. 162, 1989, pages 767 - 772
"Harrison's Principles of Internal Medicine", 1998, MCGRAW HILL
KARIMOVA ET AL., PROC NATL ACAD SCI U S A., vol. 95, no. 10, 12 May 1998 (1998-05-12), pages 5752 - 5756
SCHIAVO, G.; MATTEOLI, M.; MONTECUCCO, C.: "Neurotoxins affecting neuroexocytosis", PHYSIOL REV, vol. 80, 2000, pages 717 - 766
JOHNSON, E.A.: "Clostridial toxins as therapeutic agents: benefits of nature's most toxic proteins", ANNU REV MICROBIOL, vol. 53, 1999, pages 551 - 575, XP000943041, DOI: doi:10.1146/annurev.micro.53.1.551
AOKI, K.R.: "Botulinum toxin: a successful therapeutic protein", CURR MED CHEM, vol. 11, 2004, pages 3085 - 3092
MONTECUCCO, C.; MOLGO, J.: "Botulinal neurotoxins: revival of an old killer", CURR OPIN PHARMACOL, vol. 5, 2005, pages 274 - 279, XP004901929, DOI: doi:10.1016/j.coph.2004.12.006
LANGE, 0. ET AL.: "Neutralizing antibodies and secondary therapy failure after treatment with botulinum toxin type A: much ado about nothing?", CLIN NEUROPHARMACOL, vol. 32, 2009, pages 213 - 218
CHAPMAN, M.A.; BARRON, R.; TANIS, D.C.; GILL, C.E.; CHARLES, P.D.: "Comparison of botulinum neurotoxin preparations for the treatment of cervical dystonia", CLIN THER, vol. 29, 2007, pages 1325 - 1337, XP022230022, DOI: doi:10.1016/j.clinthera.2007.07.020
COTE, T.R.; MOHAN, A.K.; POLDER, J.A.; WALTON, M.K.; BRAUN, M.M.: "Botulinum toxin type A injections: adverse events reported to the US Food and Drug Administration in therapeutic and cosmetic cases", JAM ACADDERMATOL, vol. 53, 2005, pages 407 - 415, XP005031116
DONG, M.; TEPP, W.H.; LIU, H.; JOHNSON, E.A.; CHAPMAN, E.R.: "Mechanism of botulinum neurotoxin B and G entry into hippocampal neurons", J CELL BIOL, vol. 179, 2007, pages 1511 - 1522
PENG, L.; TEPP, W.H.; JOHNSON, E.A.; DONG, M.: "Botulinum neurotoxin D uses synaptic vesicle protein SV2 and gangliosides as receptors", PLOSPATHOG, vol. 7, 2011, pages E1002008
DONG, M. ET AL.: "Synaptotagmins I and II mediate entry of botulinum neurotoxin B into cells", J CELL BIOL, vol. 162, 2003, pages 1293 - 1303, XP002443204, DOI: doi:10.1083/jcb.200305098
NISHIKI, T. ET AL.: "Identification of protein receptor for Clostridium botulinum type B neurotoxin in rat brain synaptosomes", JBIOL CHEM, vol. 269, 1994, pages 10498 - 10503, XP002987570
RUMMEL, A.; KARNATH, T.; HENKE, T.; BIGALKE, H.; BINZ, T.: "Synaptotagmins I and II act as nerve cell receptors for botulinum neurotoxin G", JBIOL CHEM, vol. 279, 2004, pages 30865 - 30870
PENG, L. ET AL.: "Botulinum neurotoxin D-C uses synaptotagmin I/II as receptors and human synaptotagmin II is not an effective receptor for type B, D-C, and G toxins", J CELL SCI, 2012
DONG, M. ET AL.: "SV2 is the protein receptor for botulinum neurotoxin A", SCIENCE, vol. 312, 2006, pages 592 - 596, XP002423369, DOI: doi:10.1126/science.1123654
DONG, M. ET AL.: "Glycosylated SV2A and SV2B mediate the entry of botulinum neurotoxin E into neurons", MOL BIOL CELL, vol. 19, 2008, pages 5226 - 5237, XP002571458, DOI: doi:10.1091/mbc.E08-07-0765
MAHRHOLD, S.; RUMMEL, A.; BIGALKE, H.; DAVLETOV, B.; BINZ, T.: "The synaptic vesicle protein 2C mediates the uptake of botulinum neurotoxin A into phrenic nerves", FEBS LETT, vol. 580, 2006, pages 2011 - 2014, XP028030342, DOI: doi:10.1016/j.febslet.2006.02.074
RUMMEL, A. ET AL.: "Botulinum neurotoxins C, E and F bind gangliosides via a conserved binding site prior to stimulation-dependent uptake with botulinum neurotoxin F utilising the three isoforms of SV2 as second receptor", JNEUROCHEM, vol. 110, 2009, pages 1942 - 1954
FU, Z.; CHEN, C.; BARBIERI, J.T.; KIM, J.J.; BALDWIN, M.R.: "Glycosylated SV2 and gangliosides as dual receptors for botulinum neurotoxin serotype F", BIOCHEMISTRY, vol. 48, no. 563, 2009, pages 1 - 5641
MONTECUCCO, C: "How do tetanus and botulinum toxins bind to neuronal membranes?", TIBS, 1986, pages 314 - 317, XP023568661, DOI: doi:10.1016/0968-0004(86)90282-3
NISHIKI, T. ET AL.: "The high-affinity binding of Clostridium botulinum type B neurotoxin to synaptotagmin II associated with gangliosides GTlb/GDla", FEBS LETT, vol. 378, 1996, pages 253 - 257
PANG, Z.P. ET AL.: "Synaptotagmin-2 is essential for survival and contributes to Ca2+ triggering of neurotransmitter release in central and neuromuscular synapses", JNEUROSCI, vol. 26, 2006, pages 13493 - 13504, XP055412201, DOI: doi:10.1523/JNEUROSCI.3519-06.2006
STROTMEIER, J.; WILLJES, G.; BINZ, T.; RUMMEL, A.: "Human synaptotagmin-11 is not a high affinity receptor for botulinum neurotoxin B and G: increased therapeutic dosage and immunogenicity", FEBS LETT, vol. 586, 2012, pages 310 - 313, XP028455646, DOI: doi:10.1016/j.febslet.2011.12.037
CRAXTON, M: "A manual collection of Syt, Esyt, Rph3a, Rph3al, Doc2, and Dblc2 genes from 46 metazoan genomes--an open access resource for neuroscience and evolutionary biology", BMC GENOMICS, vol. 11, 2010, pages 37, XP021066160
BRIN, M.F. ET AL.: "Safety and efficacy ofNeuroBloc (botulinum toxin type B) in type A-resistant cervical dystonia", NEUROLOGY, vol. 53, 1999, pages 1431 - 1438
PAPPERT, E.J.; GERMANSON, T: "Botulinum toxin type B vs. type A in toxin-naive patients with cervical dystonia: Randomized, double-blind, noninferiority trial", MOV DISORD, vol. 23, 2008, pages 510 - 517
WANG, J. ET AL.: "Longer-acting and highly potent chimaeric inhibitors of excessive exocytosis created with domains from botulinum neurotoxin A and B", BIOCHEM J, vol. 444, 2012, pages 59 - 67, XP002690949, DOI: doi:10.1042/BJ20120100
RUMMEL, A.; MAHRHOLD, S.; BIGALKE, H.; BINZ, T.: "Exchange of the H(CC) domain mediating double receptor recognition improves the pharmacodynamic properties of botulinum neurotoxin", FEBS J, vol. 278, 2011, pages 4506 - 4515, XP002690950, DOI: doi:10.1111/J.1742-4658.2011.08196.X
KOZAKI, S. ET AL.: "Characterization of Clostridium botulinum type B neurotoxin associated with infant botulism in japan", INFECT IMMUN, vol. 66, 1998, pages 4811 - 4816
IHARA, H. ET AL.: "Sequence of the gene for Clostridium botulinum type B neurotoxin associated with infant botulism, expression of the C-terminal half of heavy chain and its binding activity", BIOCHIM BIOPHYS ACTA, vol. 1625, 2003, pages 19 - 26, XP004402176, DOI: doi:10.1016/S0167-4781(02)00537-7
RUMMEL, A.; MAHRHOLD, S.; BIGALKE, H.; BINZ, T.: "The HCC-domain of botulinum neurotoxins A and B exhibits a singular ganglioside binding site displaying serotype specific carbohydrate interaction", MOL MICROBIOL, vol. 51, 2004, pages 631 - 643, XP002363868, DOI: doi:10.1046/j.1365-2958.2003.03872.x
CHAI, Q. ET AL.: "Structural basis of cell surface receptor recognition by botulinum neurotoxin B", NATURE, vol. 444, 2006, pages 1096 - 1100, XP055404062, DOI: doi:10.1038/nature05411
JIN, R.; RUMMEL, A.; BINZ, T.; BRUNGER, A.T.: "Botulinum neurotoxin B recognizes its protein receptor with high affinity and specificity", NATURE, vol. 444, 2006, pages 1092 - 1095, XP055412605, DOI: doi:10.1038/nature05387
ARNON, S. S. ET AL.: "Botulinum toxin as a biological weapon: medical and public health management", JAMA, vol. 285, 2001, pages 1059 - 1070
MORIISHI, K. ET AL.: "Mosaic structures of neurotoxins produced from Clostridium botulinum types C and D organisms", BIOCHIM BIOPHYS ACTA, vol. 1307, 1996, pages 123 - 126, XP000857076, DOI: doi:10.1016/0167-4781(96)00006-1
HILL, K.K. ET AL.: "Genetic diversity among Botulinum Neurotoxin-producing clostridial strains", JBACTERIOL, vol. 189, 2007, pages 818 - 832, XP002508393, DOI: doi:10.1128/JB.01180-06
LALLI, G. ET AL.: "Functional characterisation of tetanus and botulinum neurotoxins binding domains", JCELL SCI, vol. 112, 1999, pages 2715 - 2724
DONG ET AL., THE JOURNAL OF CELL BIOLOGY, vol. 179, no. 7, 2007, pages 1511 - 1522
PENG ET AL., J CELL SCI., vol. 125, 2012, pages 3233 - 42
PENG ET AL., PLOSPATHOGENS, vol. 7, no. 3, 2011, pages E1002008
DONG M ET AL., THE JOURNAL OF CELL BIOLOGY, vol. 162, no. 7, 2003, pages 1293 - 1303
PENG ET AL., CELL SCI., vol. 125, 2012, pages 3233 - 42
DONG M ET AL., SCIENCE, vol. 312, no. 5773, 2006, pages 592 - 596
PENG ET AL., PLOS PATHOGENS, vol. 7, no. 3, 2011, pages EL002008
PANG, Z.P. ET AL.: "Synaptotagmin-2 is essential for survival and contributes to Ca2+ triggering of neurotransmitter release in central and neuromuscular synapses", J NEUROSCI, vol. 26, 2006, pages 13493 - 13504, XP055412201, DOI: doi:10.1523/JNEUROSCI.3519-06.2006
CRAXTON, M.: "A manual collection of Syt, Esyt, Rph3a, Rph3al, Doc2, and Dblc2 genes from 46 metazoan genomes--an open access resource for neuroscience and evolutionary biology", BMC GENOMICS, vol. 11, 2010, pages 37, XP021066160
PAPPERT, E.J.; GERMANSON, T.: "Botulinum toxin type B vs. type A in toxin-naive patients with cervical dystonia: Randomized, double-blind, noninferiority trial", MOV DISORD, vol. 23, 2008, pages 510 - 517
Attorney, Agent or Firm:
RESNICK, David S. et al. (US)
Download PDF:
Claims:
CLAIMS

1. A botulinum neurotoxin (BoNT) polypeptide comprising:

a) a protease domain;

b) a protease cleavage site;

c) a translocation domain; and

d) a modified receptor binding domain of Clostridial botulinum serotype B (B-Hc), comprising one or more substitution mutations corresponding to substitution mutations in serotype B, strain 1, selected from the group consisting of:

E1191C, E1191V, E1191L, E1191Y, S1199W, S1199E, SI 199H, Wl 178Y, Wl 178Q, Wl 178A, Wl 178S, Yl 183C, Yl 183P and combinations thereof.

2. The BoNT polypeptide of claim 1, wherein the modified (B-Hc) comprises one

substitution mutation corresponding to a substitution mutation in serotype B, strain 1, selected from the group consisting of E1191C, El 191V, E1191L, El 191 Y, S1199E, SI 199H, Wl 178Y, Wl 178Q, Wl 178A, Wl 178S, Yl 183C, and Yl 183P.

3. The BoNT polypeptide of claim 2, wherein the modified (B-Hc) comprises the

substitution mutation corresponding to El 191C in serotype B, strain 1.

4. The BoNT polypeptide of claim 2, wherein the modified (B-Hc) comprises the

substitution mutation corresponding to El 191V in serotype B, strain 1.

5. The BoNT polypeptide of claim 1, wherein the modified (B-Hc) comprises two

substitution mutations.

6. A botulinum neurotoxin (BoNT) polypeptide comprising:

a) a protease domain;

b) a protease cleavage site;

c) a translocation domain; and

d) a modified receptor binding domain of Clostridial botulinum serotype B (B-Hc), comprising two or more substitution mutations corresponding to substitution mutations in serotype B, strain 1, wherein one of the substitution mutations is selected from the group consisting of:

E1191Q, E1191M, E1191C, E1191V, E1191L, and E1191Y.

7. The BoNT polypeptide of claim 6, wherein one other of the substitution mutations corresponds to S1199W, S1199E, S1199H, SI 199Y, Wl 178Y, Wl 178Q, Wl 178 A, W1178S, Y1183C, Y1183P, S1199F or S1199L in serotype B, strain 1.

8. The BoNT polypeptide of any one of claims 5 - 7, wherein the two substitution

mutations correspond to El 191M and SI 199W, El 191M and Wl 178Q, El 191C and S1199W; E1191C and S1199Y, El 191C and Wl 178Q, El 191Q and SI 199W,

El 191 V and SI 199W, El 191 V and SI 199Y, or El 191 V and Wl 178Q, in serotype B, strain 1.

9. The BoNT polypeptide of any one of claims 5 - 8, wherein the two substitution

mutations correspond to El 191M and SI 199W in serotype B, strain 1.

10. The BoNT polypeptide of one of claims 5 - 8, wherein the two substitution mutations correspond to El 191M and Wl 178Q in serotype B, strain 1.

11. The BoNT polypeptide of one of claims 5 - 8, wherein the two substitution mutations correspond to El 191C and SI 199W in serotype B, strain 1.

12. The BoNT polypeptide of one of claims 5 - 8, wherein the two substitution mutations correspond to El 191C and SI 199Y in serotype B, strain 1.

13. The BoNT polypeptide of one of claims 5 - 8, wherein the two substitution mutations correspond to El 191C and Wl 178Q in serotype B, strain 1.

14. The BoNT polypeptide of one of claims 5 - 8, wherein the two substitution mutations correspond to El 191Q and SI 199W in serotype B, strain 1.

15. The BoNT polypeptide of one of claims 5 - 8, wherein the two substitution mutations correspond to El 191V and SI 199W in serotype B, strain 1.

16. The BoNT polypeptide of one of claims 5 - 8, wherein the two substitution mutations correspond to El 191V and SI 199 Y in serotype B, strain 1.

17. The BoNT polypeptide of one of claims 5 - 8, wherein the two substitution mutations correspond to El 191V and Wl 178Q in serotype B, strain 1.

18. The BoNT polypeptide of claim 6, wherein the modified (B-Hc) comprises three

substitution mutations.

19. The BoNT polypeptide of claim 18, wherein the three substitution mutations are at positions that correspond to E 1191 , Yl 183 and S 1199 or to E 1191 , S 1199 and W 1178 of serotype B, strain 1.

20. The BoNT polypeptide of claim 19, wherein the three substitution mutations

correspond to E1191M, S1199W and W1178Q of serotype B, strain 1.

21. A botulinum neurotoxin (BoNT) polypeptide comprising:

a) a protease domain;

b) a protease cleavage site;

c) a translocation domain; and

d) a modified receptor binding domain of Clostridial botulinum serotype B (B-Hc), comprising a substitution mutation at a position corresponding to SI 199 or SI 201 of serotype B, strain 1.

22. The BoNT polypeptide of any one of claims 1 -21, wherein the modified B-Hc is of strain 1.

23. The BoNT polypeptide of any one of claims 1-22 wherein the protease domain,

translocation domain, and protease cleavage site are from serotype selected from the group consisting of A, B, C, D, E, F, G, and combinations thereof.

24. The BoNT polypeptide of claim 23, wherein the protease domain, translocation

domain, and protease cleavage site are from serotype B, strain 1.

25. The BoNT polypeptide of claim 23, wherein the protease domain, translocation domain, and protease cleavage site are from serotype A, strain 1.

26. A polypeptide comprising a modified receptor binding domain of Clostridial

botulinum serotype B (B-Hc) comprising one or more substitution mutations corresponding to substitution mutations in serotype B, strain 1, selected from the group consisting of:

E1191C, E1191V, E1191L, E1191Y, S1199W, S1199E, SI 199H, Wl 178Y, Wl 178Q, Wl 178A, Wl 178S, Yl 183C, Yl 183P and combinations thereof.

The polypeptide of claim 26, wherein the modified (B-Hc) comprises two substitution mutations.

A polypeptide comprising a modified receptor binding domain of Clostridial botulinum serotype B (B-Hc) comprising two or more substitution mutations corresponding to substitution mutations in serotype B, strain 1, wherein one of the substitution mutations is selected from the group consisting of El 191Q, El 191M, E1191C, E1191V, E1191L, and E1191Y.

The polypeptide of claim 26, wherein one of the substitution mutations corresponds to S1199W, S1199E, S1199H, SI 199Y, Wl 178Y, Wl 178Q, Wl 178A, Wl 178S, Y1183C, Y1183P, S1199F or S1199L in serotype B, strain 1.

The polypeptide of any one of claims 27 - 29, wherein the two substitution mutations correspond to El 191M and SI 199W, El 191M and Wl 178Q, El 191C and SI 199W; E1191C and S1199Y, E1191C and W1178Q, E1191Q and S1199W, El 191V and SI 199W, El 191 V and SI 199 Y, or El 191 V and Wl 178Q, in serotype B, strain 1.

31. The polypeptide of any one of claims 27-30, wherein the two substitution mutations correspond to El 191M and SI 199W in serotype B, strain 1.

32. The polypeptide of one of claims 27-30, wherein the two substitution mutations correspond to El 191M and Wl 178Q in serotype B, strain 1.

33. The polypeptide of one of claims 27-30, wherein the two substitution mutations correspond to El 191C and SI 199W in serotype B, strain 1.

34. The polypeptide of one of claims 27-30, wherein the two substitution mutations correspond to El 191C and SI 199Y in serotype B, strain 1.

35. The polypeptide of one of claims 27-30, wherein the two substitution mutations correspond to El 191C and Wl 178Q in serotype B, strain 1.

36. The polypeptide of one of claims 27-30, wherein the two substitution mutations correspond to El 191Q and SI 199W in serotype B, strain 1.

37. The polypeptide of one of claims 27-30, wherein the two substitution mutations correspond to El 191V and SI 199W in serotype B, strain 1.

38. The polypeptide of one of claims 27-30, wherein the two substitution mutations correspond to El 191V and SI 199 Y in serotype B, strain 1.

39. The polypeptide of one of claims 27-30, wherein the two substitution mutations correspond to El 191V and Wl 178Q in serotype B, strain 1.

40. The polypeptide of claim 30, wherein the modified (B-Hc) comprises three

substitution mutations.

41. The polypeptide of claim 40, wherein the three substitution mutations are at positions that correspond to El 191, Y1183 and SI 199 or to El 191, SI 199 and Wl 178 of serotype B, strain 1.

42. The polypeptide of claim 41, wherein the three substitution mutations correspond to E1191M, S1199W and W1178Q of serotype B, strain 1.

43. The polypeptide of any one of claims 26 - 42, wherein the modified B-Hc is of strain 1.

44. A chimeric molecule comprising a first portion that is a modified receptor binding domain of Clostridial botulinum serotype B (B-Hc) linked to a second portion, wherein the modified B-Hc comprises one or more substitution mutations selected from the group consisting of:

E1191C, E1191V, E1191L, E1191Y, S1199W, S1199E, SI 199H, Wl 178Y, Wl 178Q, Wl 178A, Wl 178S, Yl 183C, Yl 183P and combinations thereof.

45. The chimeric molecule of claim 44, wherein the modified B-Hc comprises two

substitution mutations.

46. A chimeric molecule comprising a first portion that is a modified receptor binding domain of Clostridial botulinum serotype B (B-Hc) linked to a second portion, wherein the modified B-Hc comprises two or more substitution mutations

corresponding to substitution mutations in serotype B, strain 1, wherein one of the substitution mutations is selected from the group consisting of:

E1191Q, E1191M, E1191C, E1191V, E1191L, and E1191Y.

47. The chimeric molecule of claim 46, wherein one of the substitution mutations

corresponds to S1199W, S1199E, S1199H, SI 199Y, Wl 178Y, Wl 178Q, Wl 178 A, W1178S, Y1183C, Y1183P, S1199F or S1199L in serotype B, strain 1.

48. The chimeric molecule of any one of claims 45 - 47, wherein the two substitution mutations correspond to El 191M and SI 199W, El 191M and Wl 178Q, El 191C and S1199W; E1191C and S1199Y, El 191C and Wl 178Q, El 191Q and SI 199W,

El 191 V and SI 199W, El 191 V and SI 199Y, or El 191 V and Wl 178Q, in serotype B, strain 1.

49. The chimeric molecule of any one of claims 45 - 47, wherein the two substitution mutations correspond to El 191M and SI 199W in serotype B, strain 1.

50. The chimeric molecule of any one of claims 45 - 47, wherein the two substitution mutations correspond to El 191M and Wl 178Q in serotype B, strain 1.

51. The chimeric molecule of any one of claims 45 - 47, wherein the two substitution mutations correspond to El 191C and SI 199W in serotype B, strain 1.

52. The chimeric molecule of any one of claims 45 - 47, wherein the two substitution mutations correspond to El 191C and SI 199 Y in serotype B, strain 1.

53. The chimeric molecule of any one of claims 45 - 47, wherein the two substitution mutations correspond to El 191C and Wl 178Q in serotype B, strain 1.

54. The chimeric molecule of any one of claims 45 - 47, wherein the two substitution mutations correspond to El 191Q and SI 199W in serotype B, strain 1.

55. The chimeric molecule of any one of claims 45 - 47, wherein the two substitution mutations correspond to El 191V and SI 199W in serotype B, strain 1.

56. The chimeric molecule of any one of claims 45 - 47, wherein the two substitution mutations correspond to El 191V and SI 199 Y in serotype B, strain 1.

57. The chimeric molecule of any one of claims 45 - 47, wherein the two substitution mutations correspond to El 191V and Wl 178Q in serotype B, strain 1.

58. The chimeric molecule of one of claim 45 or 46, wherein the modified (B-Hc)

comprises three substitution mutations.

59. The chimeric molecule of claim 58, wherein the three substitution mutations are at positions that correspond to E 1191 , Yl 183 and S 1199 or to E 1191 , S 1199 and W 1178 of serotype B, strain 1.

60. The chimeric molecule of claim 59, wherein the three substitution mutations

correspond to E1191M, S1199W and W1178Q of serotype B, strain 1.

61. The chimeric molecule of any one of claims 44- 60, wherein the modified B-Hc is of strain 1.

62. The chimeric molecule of any one of claims 44-61, wherein the first portion and the second portion are linked covalently.

63. The chimeric molecule of any one of claims 46-61, wherein the first portion and the second portion are linked non-covalently.

64. The chimeric molecule of any one of claims 44-61 wherein the second portion is selected from the group consisting of a small molecule, a nucleic acid, a short polypeptide and a protein.

65. The chimeric molecule of claim 64, wherein the second portion is a bioactive

molecule.

66. The chimeric molecule of claim 64 or 65, wherein the second portion is a therapeutic polypeptide or non-polypeptide drug.

67. The BoNT polypeptide, polypeptide or chimeric molecule of any one of claims 1-66 that exhibits significantly enhanced binding of the modified B-Hc to human Sytll and/or significantly reduced binding of the modified B-Hc to human Syt I as compared to an identical molecule lacking the substitution mutation(s).

68. The BoNT polypeptide, polypeptide or chimeric molecule of any one of claims 1- 66 wherein the substitution mutation produces significantly enhanced binding to human Sytll and/or significantly enhanced binding to human Syt I as compared to an identical molecule lacking the substitution mutation(s).

69. A nucleic acid comprising a nucleotide sequence that encodes the polypeptide or chimeric molecule of any one of claims 1- 68.

70. A nucleic acid vector comprising the nucleic acid of claim 69. A cell comprising the nucleic acid vector of claim 70 or the nucleic acid of claim 69.

A cell expressing the polypeptide or chimeric molecule of any one of claims 1-

A pharmaceutical composition comprising the botulinum neurotoxin (BoNT) polypeptide of any one of claims 1-25, 67 or 68, or the chimeric molecule of any one of claims 44-66, or the nucleic acid vector of claim 70 or the nucleic acid of claim 69.

74. The pharmaceutical composition of claim 73, further comprising a pharmaceutically acceptable excipient.

75. A kit comprising a pharmaceutical composition of claim 73 or 74 and directions for therapeutic administration of the pharmaceutical composition.

A method to produce a botulinum neurotoxin (BoNT) polypeptide, the method comprising the steps of culturing the cell of claim 72 under conditions wherein said BoNT polypeptide is produced.

The method of claim 76 further comprising one or more of the following

- recovering the BoNT polypeptide from the culture,

- purifying the BoNT polypeptide,

- activating the BoNT polypeptide, and/or

- formulating the BoNT polypeptide.

A method for treating a condition associated with unwanted neuronal activity comprising administering a therapeutically effective amount of the BoNT polypeptide of any one of claims 1-25, 67 or 68 to a subject to thereby contact one or more neurons exhibiting unwanted neuronal activity, to thereby treat the condition.

The method of claim 78, wherein the condition is selected from the group consisting of , spasmodic dysphonia, spasmodic torticollis, laryngeal dystonia, oromandibular dysphonia, lingual dystonia, cervical dystonia, focal hand dystonia, blepharospasm, strabismus, hemifacial spasm, eyelid disorder, cerebral palsy, focal spasticity and other voice disorders, spasmodic colitis, neurogenic bladder, anismus, limb spasticity, tics, tremors, bruxism, anal fissure, achalasia, dysphagia and other muscle tone disorders and other disorders characterized by involuntary movements of muscle groups, lacrimation, hyperhydrosis, excessive salivation, excessive gastrointestinal secretions, secretory disorders, pain from muscle spasms, headache pain, migraine, and dermatological or aesthetic/cosmetic conditions.

80. The botulinum neurotoxin (BoNT) polypeptide of any one of claims 1-25, 67 or 68, the pharmaceutical composition of one of claims 73 or 74 or the chimeric molecule of any one of claims 44-66, or the polypeptide of any one of claims 26-43, for use in medicine.

81. The botulinum neurotoxin (BoNT) polypeptide of any one of claims 1-25, 67 or 68, the pharmaceutical composition of one of claims 73 or 74, or the chimeric molecule of any one of claims 44-66, or the polypeptide of any one of claims 67-43, for use in treating a condition associated with unwanted neuronal activity.

82. A method for identifying a modified receptor binding domain of botulinum

neurotoxin for binding to a receptor comprising;

a) expressing the modified receptor binding domain as a first fusion protein with T18 subunit of bacterial adenylate cyclase in a 2-hybrid assay, and expressing the receptor as a second fusion protein with T25 subunit of bacterial adenylate cyclase in the 2-hybrid assay;

b) analyzing a clonal E. coli colony that expresses both the first and second fusion protein for the presence of a positive indication above a negative control; and

c) identifying the modified receptor binding domain expressed by the colony exhibiting the positive indication as binding to the receptor.

83. The method of claim 82 wherein the positive indication is color development.

84. The method of any one of claims 82 - 83, further comprising the step of analyzing the colony for the positive indication against a weakly positive control, and further identifying the modified receptor binding domain expressed by the colony exhibiting above the weakly positive control as binding to the receptor with high affinity.

85. The method of claim 82-84 that is performed with a library of modified receptor binding domains each expressed within respective colonies.

86. The method of any one of claims 82-85 wherein the receptor is human.

87. The method of any one of claims 82-86 wherein the modified receptor binding

domain comprises one or more substitution mutations, wherein one of the substitution mutations corresponds to a mutation in serotype B, strain 1, selected from the group consisting of: El 191M, El 191 Q, El 191C, El 191 V, El 191L, El 191 Y, SI 199W, S1199E, S1199H, S1199F, SI 199L Wl 178Y, Wl 178Q, Wl 178A, Wl 178S, Yl 183C, Y1183P and S1199Y.

Description:
ENGINEERED BOTULINUM NEUROTOXIN

RELATED APPLICATIONS

[0001] This Application claims the benefit under 35 U.S.C. § 119(e) of U. S. Provisional Application serial number 62/138,818, filed March 26, 2015, the contents of which are incorporated herein by reference in their entirety.

GOVERNMENTAL SUPPORT

[0002] This invention was made with Government support under NIH NCRR RR000168 awarded by the National Institute of Health. The Government has certain rights in the invention.

FIELD OF THE INVENTION

[0003] The present invention relates to the field of modified neurotoxins and their use for treating medical and/or aesthetic disorders.

BACKGROUND OF THE INVENTION

[0004] Botulinum neurotoxins are a family of bacterial toxins, including seven major serotypes (BoNT/A-G) \ These toxins act by blocking neurotransmitter release from neurons, thus paralyzing animals and humans. In recent years, BoNTs have been widely used to treat a growing list of medical conditions: local injections of minute amount of toxins can attenuate neuronal activity in targeted regions, which can be beneficial in many medical conditions as well as for cosmetic purposes 2"4 .

[0005] BoNT/A and BoNT/B are the only two BoNTs that are currently FDA-approved for use in humans 2-4 . These are toxins purified from bacteria without any sequence modifications (defined as wild type, WT). As the application of BoNTs grows, limitations and adverse effects have been reported. The major limitation is the generation of neutralizing antibodies in patients, which renders future treatment ineffective 5 . Termination of BoNT usage often leaves patients with no other effective ways to treat/relieve their disorders. The possibility of antibody responses is directly related to both toxin doses and the frequency of injection 5 . Therefore, this limitation mainly occurs in treating muscle spasms, which involves relatively high doses of toxins. Consistently, antibody responses have not been observed in cosmetic applications, which use extremely low toxin doses 5 . [0006] The major adverse effects are also often associated with treating muscle spasms, but not cosmetic applications. This is because the adverse effects are largely due to diffusion of toxins to other regions of the body and the possibility of toxin diffusion is directly related to injected doses. The adverse effects ranges from transient non-serious events such as ptosis and diplopia to life-threatening events even death 6 ' 7 . In a petition letter filed in 2008 by Dr. Sidney Wolfe to FDA, a total of 180 serious adverse events, including 16 deaths have been documented. As a result, FDA now requires the "Black box warning" on all BoNT products, highlighting the risk of the spread of toxins, following similar warnings issued by the European Union.

[0007] Because both the generation of neutralizing antibodies and toxin diffusion are directly related to injected doses, lowering toxin doses (while maintaining the same levels of toxin activity) is highly desired, which means the efficacy of individual toxin molecules has to be enhanced. Such modified BoNTs with improved specificity for neurons will also reduce any potential off-target effects due to non-specific entry into other cell types.

[0008] BoNTs target and enter neurons by binding to their specific receptors through their receptor binding domains, which are well-defined in the literature (BoNT-Hc, Fig. 1 A, B) \ Receptor binding dictates the efficacy and specificity of BoNTs to recognize neurons.

Improving the receptor binding ability of BoNTs will enhance their efficacy and specificity to target neurons. The receptors for most BoNTs have been identified (Fig. 1C). BoNT/B, D-C, and G share two homologous synaptic vesicle proteins synaptotagmin I and II (Syt VII) as their receptors 8"13 , while BoNT/ A, E, D, and F use another synaptic vesicle protein SV2 9 14" 18 . In addition to protein receptors, all BoNTs require lipid co-receptor gangliosides (Fig. ID), which are abundant on neuronal surfaces 19 . Among the two Syt isoforms Syt II has ~10-fold higher binding affinity for BoNT/B than Syt I and is also the dominant isoform expressed in motor nerve terminals, which are the targeted neurons for BoNTs (Fig. 2A) 20 ' 21 . Therefore, Syt II is considered the major toxin receptor for BoNT/B, D-C, and G, while Syt I is a minor toxin receptor at motor nerve terminals. This is the case in rodents and likely in the majority of mammals.

[0009] One may argue that BoNTs already have high specificity to neurons, and ask is it possible to further improve their binding to neurons? The answer is a "Yes" for humans, at least in part in light of the recent discovery that the human Syt II has greatly diminished binding and function as the receptor for BoNT/B due to a unique amino acid change from rodent (rat/mouse) Syt II within the toxin binding site 13 ' 22 . This is a change from

phenylalanine (F) to leucine (L) at position 54 (mouse Syt II sequence) (Fig. 2B). Sequence alignments have revealed that phenylalanine at this position is highly conserved in both Syt I and Syt II across vertebrates, including platypus, fish, rodents, and monkeys 23 . Only human and chimpanzee Syt II contains leucine at this position. As a result of this residue change, human and chimpanzee Syt II has greatly diminished binding to BoNT/B, D-C, and G (Fig. 2C) and is significantly less efficient in mediating the entry of BoNT/B (Fig. 2D), as compared to mouse Syt II. Since human and chimpanzee Syt I still contains phenylalanine at the same position and can bind BoNT/B, D-C, and G (Fig. 2E), the high affinity receptor for BoNT/B, D-C, and G in humans is restricted to the minor receptor Syt I. These findings provide an explanation for the clinical observations that a much higher dose of BoNT/B than BoNT/A (which binds a different receptor) is needed to achieve the same levels of therapeutic effects in patients 24 ' 25 . Previously these observations were attributed to other reasons, such as the percentage of active neurotoxin in the preparations used. The recent observations of such binding differences of BoNT/B to human Syt II versus Syt II of other species suggests that different residues of BoNT/B may be involved in binding to human Syt II. As such, sequence modification to BoNT/B that is expected to affect binding to rodent Sytll may have unpredictable effects on BoNT/B binding to human Syt II.

[0010] Thus there is a great need to improve binding of BoNT/B to human receptor Syt II, as a way to increase its efficacy and specificity to target human neurons and decrease the toxin doses needed in therapeutic applications.

SUMMARY OF THE INVENTION

[0011] One aspect of the invention relates to a botulinum neurotoxin (BoNT) polypeptide comprising a protease domain, a protease cleavage site, a translocation domain, and a modified receptor binding domain of Clostridial botulinum serotype B (B-H c ), comprising one or more substitution mutations corresponding to substitution mutations in serotype B, strain 1, selected from the group consisting of El 191C, El 191 V, El 191L, El 191 Y,

S1199W, S1199E, S1199H, Wl 178Y, Wl 178Q, Wl 178A, Wl 178S, Yl 183C, Yl 183P, and combinations thereof. In one embodiment, the modified (B-H c ) comprises one substitution mutation corresponding to a substitution mutation in serotype B, strain 1, selected from the group consisting of El 191C, El 191 V, El 191L, El 191 Y, SI 199E, SI 199H, Wl 178Y, Wl 178Q, Wl 178A, Wl 178S, Yl 183C, and Yl 183P. In one embodiment of the

polypeptides described herein the modified (B-H c ) comprises the substitution mutation corresponding to El 191C in serotype B, strain 1. In one embodiment of the polypeptides described herein the modified (B-H c ) comprises the substitution mutation corresponding to El 191V in serotype B, strain 1. In one embodiment of the polypeptides described herein the modified (B-H c ) comprises two substitution mutations.

[0012] Another aspect of the invention relates to a botulinum neurotoxin (BoNT) polypeptide comprising a protease domain, a protease cleavage site, a translocation domain, and a modified receptor binding domain of Clostridial botulinum serotype B (B-H c ), comprising two or more substitution mutations corresponding to substitution mutations in serotype B, strain 1, wherein one of the substitution mutations is selected from the group consisting of El 191Q, El 191M, El 191C, El 191 V, El 191L, and El 191 Y. In one embodiment, one other of the substitution mutations corresponds to SI 199W, SI 199E, SI 199H, SI 199Y, Wl 178Y, W1178Q, W1178A, W1178S, Y1183C, Y1183P, S1199F or S1199L in serotype B, strain 1. In one embodiment of the polypeptides described herein the two substitution mutations correspond to El 191M and SI 199W, El 191M and Wl 178Q, El 191C and SI 199W; El 191C and SI 199Y, El 191C and Wl 178Q, El 191Q and SI 199W, El 191 V and SI 199W, El 191 V and SI 199 Y, or El 191V and W1178Q, in serotype B, strain 1. In one embodiment of the polypeptides described herein the two substitution mutations correspond to El 191M and SI 199W in serotype B, strain 1. In one embodiment of the polypeptides described herein the two substitution mutations correspond to El 191M and Wl 178Q in serotype B, strain 1. In one embodiment of the polypeptides described herein the two substitution mutations correspond to El 191C and SI 199W in serotype B, strain 1. In one embodiment of the polypeptides described herein the two substitution mutations correspond to El 191C and SI 199 Y in serotype B, strain 1. In one embodiment of the polypeptides described herein the two substitution mutations correspond to El 191C and Wl 178Q in serotype B, strain 1. In one embodiment of the polypeptides described herein the two substitution mutations correspond to El 191Q and SI 199W in serotype B, strain 1. In one embodiment of the polypeptides described herein the two substitution mutations correspond to El 191 V and SI 199W in serotype B, strain 1. In one embodiment of the polypeptides described herein the two substitution mutations correspond to El 191V and SI 199Y in serotype B, strain 1. In one embodiment of the polypeptides described herein the two substitution mutations correspond to El 191V and Wl 178Q in serotype B, strain 1. In one embodiment of the polypeptides described herein the modified (B-H c ) comprises three substitution mutations. In one embodiment of the polypeptides described herein the three substitution mutations are at positions that correspond to El 191, Y1183 and SI 199 or to El 191, SI 199 and Wl 178 of serotype B, strain 1. In one embodiment of the polypeptides described herein the three substitution mutations correspond to El 191M, SI 199W and Wl 178Q of serotype B, strain 1. [0013] Another aspect of the invention relates to a botulinum neurotoxin (BoNT) polypeptide comprising a protease domain, a protease cleavage site, a translocation domain, and a modified receptor binding domain of Clostridial botulinum serotype B (B-H c ), comprising a substitution mutation at a position corresponding to SI 199 or S1201 of serotype B, strain 1.

[0014] In one embodiment of any of the polypeptides described herein the modified B-H c is of strain 1. In one embodiment of any of the polypeptides described herein the protease domain, translocation domain, and protease cleavage site are from serotype selected from the group consisting of A, B, C, D, E, F, G, and combinations thereof. In one embodiment of any of the polypeptides described herein the protease domain, translocation domain, and protease cleavage site are from serotype B, strain 1. In one embodiment of any of the polypeptides described herein the protease domain, translocation domain, and protease cleavage site are from serotype A, strain 1. In one embodiment of any of the polypeptides described herein, the modified B-Hc is not of strain B4.

[0015] Another aspect of the invention relates to a polypeptide comprising a modified receptor binding domain of Clostridial botulinum serotype B (B-H c ) comprising one or more substitution mutations corresponding to substitution mutations in serotype B, strain 1, selected from the group consisting of E1191C, El 191V, E1191L, El 191 Y, S1199W, SI 199E, SI 199H, Wl 178Y, Wl 178Q, Wl 178A, Wl 178S, Yl 183C, Yl 183P, and combinations thereof. In one embodiment, the modified (B-H c ) comprises two substitution mutations.

[0016] Another aspect of the invention relates to a polypeptide comprising a modified receptor binding domain of Clostridial botulinum serotype B (B-H c ) comprising two or more substitution mutations corresponding to substitution mutations in serotype B, strain 1, wherein one of the substitution mutations is selected from the group consisting of El 191Q, El 191M, El 191C, El 191 V, El 191L, and El 191 Y. In one embodiment of the polypeptides described herein one of the substitution mutations corresponds to SI 199W, SI 199E, SI 199H, S1199Y, W1178Y, W1178Q, W1178A, W1178S, Y1183C, Y1183P, S1199F or S1199L in serotype B, strain 1. In one embodiment of the polypeptides described herein the two substitution mutations correspond to El 191M and SI 199W, El 191M and Wl 178Q, El 191C and S1199W; E1191C and S1199Y, E1191C and W1178Q, E1191Q and S1199W, El 191V and SI 199W, El 191 V and SI 199 Y, or El 191 V and Wl 178Q, in serotype B, strain 1. In one embodiment of the polypeptides described herein the two substitution mutations correspond to El 191M and SI 199W in serotype B, strain 1. In one embodiment of the polypeptides described herein the two substitution mutations correspond to El 191M and Wl 178Q in serotype B, strain 1. In one embodiment of the polypeptides described herein the two substitution mutations correspond to El 191C and SI 199W in serotype B, strain 1. In one embodiment of the polypeptides described herein the two substitution mutations correspond to El 191C and SI 199Y in serotype B, strain 1. In one embodiment of the polypeptides described herein the two substitution mutations correspond to El 191C and Wl 178Q in serotype B, strain. In one embodiment of the polypeptides described herein the two substitution mutations correspond to El 191Q and SI 199W in serotype B, strain 1. In one embodiment of the polypeptides described herein the two substitution mutations correspond to El 191V and SI 199W in serotype B, strain 1. In one embodiment of the polypeptides described herein the two substitution mutations correspond to El 191 V and SI 199Y in serotype B, strain 1. In one embodiment of the polypeptides described herein the two substitution mutations correspond to El 191V and Wl 178Q in serotype B, strain 1. In one embodiment of the polypeptides described herein the modified (B-H c ) comprises three substitution mutations. In one embodiment of the polypeptides described herein the three substitution mutations are at positions that correspond to El 191, Yl 183 and SI 199 or to El 191, SI 199 and Wl 178 of serotype B, strain 1. In one embodiment of the polypeptides described herein the three substitution mutations correspond to El 191M, SI 199W and Wl 178Q of serotype B, strain 1. In one embodiment of the polypeptides described herein the modified B-H c is of strain 1. In one embodiment of the polypeptides described herein the modified B-Hc is not of strain 4. In one embodiment, the modified B-Hc is not of strain 3, 7 or 8.

[0017] Another aspect of the invention relates to a chimeric molecule comprising a first portion that is a modified receptor binding domain of Clostridial botulinum serotype B (B- H c ) linked to a second portion, wherein the modified B-H c comprises one or more substitution mutations selected from the group consisting of El 191C, El 191 V, El 191L, El 191 Y, S1199W, S1199E, S1199H, Wl 178Y, Wl 178Q, Wl 178A, Wl 178S, Yl 183C, Yl 183P, and combinations thereof. In one embodiment the modified B-H c comprises two substitution mutations.

[0018] Another aspect of the invention relates to a chimeric molecule comprising a first portion that is a modified receptor binding domain of Clostridial botulinum serotype B (B- H c ) linked to a second portion, wherein the modified B-H c comprises two or more

substitution mutations corresponding to substitution mutations in serotype B, strain 1, wherein one of the substitution mutations is selected from the group consisting of El 191Q, El 191M, El 191C, El 191 V, El 191L, and El 191 Y. In one embodiment of the chimeric molecules described herein one of the substitution mutations corresponds to SI 199W, S1199E, S1199H, SI 199Y, Wl 178Y, Wl 178Q, Wl 178A, Wl 178S, Y1183C, Y1183P, SI 199F or SI 199L in serotype B, strain 1. In one embodiment of the chimeric molecules described herein the two substitution mutations correspond to El 191M and SI 199W, E1191M and W1178Q, E1191C and S1199W; E1191C and S1199Y, E1191C and W1178Q, E1191Q and SI 199W, El 191V and SI 199W, El 191V and S1199Y, or El 191V and

Wl 178Q, in serotype B, strain 1. In one embodiment of the chimeric molecules described herein the two substitution mutations correspond to El 191M and SI 199W in serotype B, strain l .In one embodiment of the chimeric molecules described herein the two substitution mutations correspond to El 191M and Wl 178Q in serotype B, strain 1. In one embodiment of the chimeric molecules described herein the two substitution mutations correspond to El 191C and SI 199W in serotype B, strain 1. In one embodiment of the chimeric molecules described herein the two substitution mutations correspond to El 191C and SI 199Y in serotype B, strain 1. In one embodiment of the chimeric molecules described herein the two substitution mutations correspond to El 191C and Wl 178Q in serotype B, strain 1. In one embodiment of the chimeric molecules described herein the two substitution mutations correspond to El 191Q and SI 199W in serotype B, strain 1. In one embodiment of the chimeric molecules described herein the two substitution mutations correspond to El 191 V and SI 199W in serotype B, strain 1. In one embodiment of the chimeric molecules described herein the two substitution mutations correspond to El 191 V and SI 199Y in serotype B, strain 1. In one embodiment of the chimeric molecules described herein the two substitution mutations correspond to El 191V and Wl 178Q in serotype B, strain 1. In one embodiment of the chimeric molecules described herein the modified (B-H c ) comprises three substitution mutations. In one embodiment of the chimeric molecules described herein the three substitution mutations are at positions that correspond to El 191, Yl 183 and SI 199 or to El 191, S 1199 and Wl 178 of serotype B, strain 1. In one embodiment of the chimeric molecules described herein the three substitution mutations correspond to El 191M, SI 199W and Wl 178Q of serotype B, strain 1.

[0019] In one embodiment of any of the chimeric molecules described herein the modified B- H c is of strain 1. In one embodiment of any of the chimeric molecules described herein, the modified B-Hc is not of strain 4. In one embodiment, the modified B-Hc is not of strain 3, 7 or 8.

[0020] In one embodiment of any of the chimeric molecules described herein the first portion and the second portion are linked covalently. In one embodiment of any of the chimeric molecules described herein the first portion and the second portion are linked non-covalently. In one embodiment of any of the chimeric molecules described herein the second portion is selected from the group consisting of a small molecule, a nucleic acid, a short polypeptide and a protein. In one embodiment of any of the chimeric molecules described herein the second portion is a bioactive molecule. In one embodiment of any of the chimeric molecules described herein the second portion is a therapeutic polypeptide or non-polypeptide drug.

[0021] Aspects of the invention further relate to any of the BoNT polypeptides, polypeptides or chimeric molecules described herein that exhibits significantly enhanced binding of the modified B-Hc to human Sytll and/or significantly reduced binding of the modified B-Hc to human Syt I as compared to an identical molecule lacking the substitution mutation(s).

[0022] Aspects of the invention further relate to any of the BoNT polypeptides, polypeptides or chimeric molecules described herein wherein the substitution mutation produces significantly enhanced binding to human Sytll and/or significantly enhanced binding to human Syt I as compared to an identical molecule lacking the substitution mutation(s).

[0023] Another aspect of the invention relates to a nucleic acid comprising a nucleotide sequence that encodes the BoNT polypeptide, polypeptide or chimeric molecule described herein. Another aspect of the invention relates to a nucleic acid vector comprising the nucleic acid comprising a nucleotide sequence that encodes the BoNT polypeptide, polypeptide or chimeric molecule described herein. Another aspect of the invention relates to a cell comprising the nucleic acid comprising a nucleotide sequence that encodes the BoNT polypeptide, polypeptide or chimeric molecule described herein, or the nucleic acid vector comprising the nucleic acid.

[0024] Another aspect of the invention relates to a cell expressing any of the BoNT polypeptides, polypeptides or chimeric molecules described herein.

[0025] Another aspect of the invention relates to a pharmaceutical composition comprising any of the botulinum neurotoxin (BoNT) polypeptides described herein.

[0026] Another aspect of the invention relates to a pharmaceutical composition comprising any of the polypeptides described herein.

[0027] Another aspect of the invention relates to a pharmaceutical composition comprising any of the chimeric molecules described herein.

[0028] Another aspect of the invention relates to a pharmaceutical composition comprising any of the nucleic acids or nucleic acid vectors described herein.

[0029] In one embodiment of any of the pharmaceutical compositions described herein the pharmaceutical composition further comprises a pharmaceutically acceptable excipient. [0030] Another aspect of the invention relates to a kit comprising any of the pharmaceutical compositions described herein and directions for therapeutic administration of the

pharmaceutical composition.

[0031] Another aspect of the invention relates to a method to produce any of the botulinum neurotoxin (BoNT) polypeptides described herein, the method comprising the steps of culturing the cell that expresses the BoNT polypeptide under conditions wherein said BoNT polypeptide is produced. In one embodiment, the method further comprises one or more of the steps of recovering the BoNT polypeptide from the culture, purifying the BoNT polypeptide, activating the BoNT polypeptide, and/or formulating the BoNT polypeptide.

[0032] Another aspect of the invention relates to a method for treating a condition associated with unwanted neuronal activity comprising administering a therapeutically effective amount of the BoNT polypeptide described herein to a subject to thereby contact one or more neurons' exhibiting unwanted neuronal activity, to thereby treat the condition. In one embodiment, the condition is selected from the group consisting of , spasmodic dysphonia, spasmodic torticollis, laryngeal dystonia, oromandibular dysphonia, lingual dystonia, cervical dystonia, focal hand dystonia, blepharospasm, strabismus, hemifacial spasm, eyelid disorder, cerebral palsy, focal spasticity and other voice disorders, spasmodic colitis, neurogenic bladder (i.e. all diseases involving urinary incontinence, such as e.g. neurogenic detrusor overactivity or idiopathic overactive bladder), anismus, limb spasticity, tics, tremors, bruxism, anal fissure, achalasia, dysphagia and other muscle tone disorders and other disorders characterized by involuntary movements of muscle groups, lacrimation,

hyperhydrosis, excessive salivation, excessive gastrointestinal secretions, secretory disorders, pain from muscle spasms, neuropathic pain, inflammatory pain, headache pain (such as e.g., migraine), itch (pruritis), acne, and dermatological or aesthetic/cosmetic conditions.

[0033] Another aspect of the invention relates to any of the botulinum neurotoxin (BoNT) polypeptides or the pharmaceutical compositions, or the chimeric molecules, or the polypeptides described herein for use in medicine.

[0034] Another aspect of the invention relates to any of the botulinum neurotoxin (BoNT) polypeptides, or the pharmaceutical compositions, or the chimeric molecules, or the polypeptides described herein, for use in treating a condition associated with unwanted neuronal activity.

[0035] Another aspect of the invention relates to a method for identifying a modified receptor binding domain of botulinum neurotoxin for binding to a receptor comprising expressing the modified receptor binding domain as a first fusion protein with T18 subunit of bacterial adenylate cyclase in a 2-hybrid assay, and expressing the receptor as a second fusion protein with T25 subunit of bacterial adenylate cyclase in the 2-hybrid assay, analyzing a clonal E. coli colony that expresses both the first and second fusion protein for the presence of a positive indication above a negative control, and identifying the modified receptor binding domain expressed by the colony exhibiting the positive indication as binding to the receptor. In one embodiment, the positive indication is color development. In one embodiment of the methods described herein, the method further comprises the step of analyzing the colony for the positive indication against a weakly positive control, and further identifying the modified receptor binding domain expressed by the colony exhibiting the positive indication above the weakly positive control as binding to the receptor with high affinity. In one embodiment of the methods described herein, the method is performed with a library of modified receptor binding domains each expressed within respective colonies. In one embodiment of the methods described herein, the receptor is human. In one embodiment of the methods described herein, the modified receptor binding domain comprises one or more substitution mutations, wherein one of the substitution mutations corresponds to a mutation in serotype B, strain 1, selected from the group consisting of: El 191M, El 191 Q, El 191C, E1191V, E1191L, E1191Y, S1199W, S1199E, S1199H, S1199F, SI 199L Wl 178Y, Wl 178Q, Wl 178A, Wl 178S, Yl 183C, Yl 183P and SI 199Y.

BRIEF DESCRIPTION OF THE DRAWINGS

[0036] Figure 1 A- Figure ID (published data for background) shows schematic models for how BoNTs target neurons (Figure 1 A), their overall protein structure (Figure IB), a list of identified receptors (Figure 1C), and the structural model for BoNT/B binding to its receptors Syt and gangliosides (Figure ID). (Figure 1 A) A schematic view of BoNT actions: BoNTs recognize neurons by binding to their specific receptors (step 1), enter neurons via receptor- mediated endocytosis (step 2), the light chains of BoNTs then translocate across endosomal membranes into the cytosol (step 3), where these light chains act as proteases to cleave target host proteins (step 4). Figure 1 A is adapted from Arnon, S. et al, JAMA, 285:1059, 2001 24 . (Figure IB) BoNTs are composed of a light chain and a heavy chain, connected via a disulfide bond. The heavy chain can be further divided into two domains: the translocation domain (H N ) and the receptor binding domain (He). These functional domains are switchable between different BoNTs. For instance, BoNT/B-Hc can be used to replace BoNT/A-Hc to generate chimeric toxins. (Figure 1C) A list of identified toxins receptors. (Figure ID) A structural model showing binding of BoNT/B to its protein receptor, Syt (I/II), as well as its lipid co-receptor, gangliosides, on the cell surface. Figure ID is adapted from Chai et al, Nature, 444:1096, 2006 31 .

[0037] Figure 2A - Figure 2D show prior data adapted from published literatures showing indicating that human Syt II is not an effective receptor for BoNT/B, D-C, and G. (A) Human Syt II differs from mouse/rat Syt II by a single residue within the toxin binding site (residue 54 in mouse Syt II, 51 in human Syt II). (Sytll (mouse) shown is SEQ ID NO: 1). (Figure 2B) Glutathione ^-transferase (GST) tagged recombinant mouse Syt II 1-87 (m-Syt II) and a mouse Syt II 1-87 mutant mimicking human Syt II (F54L, here and thereafter designed as h- Syt II to simplify the text) were immobilized on glutathione-sepharose beads, and were used to pull down BoNT/B, BoNT/D-C, or BoNT/G, with or without the presence of ganglioside (Gangl). All three toxins bind to m-Syt II 1-87, but not h-Syt II in the pull-down assays. (Figure 2C) Cultured rat hippocampal neurons only express Syt I but not Syt II 8 . Therefore, knocking down (KD) Syt I generates neurons with no endogenous toxin receptors. Full- length m-Syt II and h-Syt II were then expressed in Syt I KD hippocampal neurons, and these neurons were exposed to BoNT/B (20 nM, 5 min exposure, 24 hours incubation) or BoNT/D- C ( 0.3 nM, 5 min exposure, 6 hours incubation) or BoNT/G (40 nM, 5 min exposure, 24 hours incubation). h-Syt II was found significantly less efficient in mediating the entry of BoNT/B, BoNT/D-C, and BoNT/G into Syt I KD neurons as compared to wild type mouse Syt II, as evidenced by the degrees of cleavage of toxin substrate synaptobrevin (Syb).

(Figure 2D) Rat Syt 1 1-83 and human Syt 1 1-80 were used to pull down BoNT/B, BoNT/D- C, and BoNT/G, as described in panel C. Human Syt I mediated similar levels of toxin binding as rat Syt I did for all three toxins. This figure is adapted from our recent publication: Peng et al, J. Cell Science, 2012 13 .

[0038] Figure 3 A- Figure 3B shows residues located in the Syt II binding interface on BoNT/B-Hc, which are candidates for site-directed mutagenesis to modify the binding affinity. (Figure 3 A) A Close-up view of binding interface between BoNT/B-H c and mouse Syt II is shown here. Residues in BoNT/B-Hc that contribute to the binding are marked in bold, whereas residues in Syt II are not bold. Residue F54 in Syt II, in the middle and shown in slightly larger font size, changes to residue L in human Syt II. (Figure 3B) A list of 19 key residues in BoNT/B that form the binding pocket of the Syt II binding domain in BoNT/B. These residues are candidates for site-directed mutagenesis.

[0039] Figure 4A- Figure 4B are illustrations of the bacterial adenylate cyclase two hybrids (BACTH) system that is used to screen BoNT/B-H c mutants for their ability to bind human Syt II. (Figure 4 A) A pool of BoNT/B-H c mutants that covers all 20 different amino acids at the selected position is generated by PCR amplification with primers containing random trinucleotides (NNN) at the selected site. A total of 19 distinct pools were generated for all 19 residues selected in Figure 3B. (Figure 4B) A schematic illustration of the BACTH system. Briefly, the bacterial adenylate cyclase is split to two domains, designated as T25 and T18. T25 is fused to human Syt II (residues 1-87) that contains the toxin binding site, whereas T18 is fused to BoNT/B-H c generated as described in Figure 4A. These two fusion proteins are encoded on two separated plasmids and co-transformed into the same bacteria. Binding of mutated ΒοΝΤ/Β-¾ to human Syt II brings together T25 and T18 domains in bacteria, which restores the activity of the bacterial adenylate cyclase. This leads to production of cAMP in bacteria, which activates CAP protein and triggers expression of the reporter gene lacZ that encodes β-galactosidase. The activity of β-galactosidase leads to blue colonies on X-gal plates that can be easily identified. Plasmids that encode BoNT/B-Hc can be then extracted from blue colonies and sequenced to identify the specific mutations in BoNT/B-H c .

[0040] Figure 5 A - Figure 5B are tables that summarize the results from the BACTH screening of ΒοΝΤ/Β-¾ mutants. (Figure 5 A) List of total colonies and the number of blue colonies for each pool of ΒοΝΤ/Β-¾ mutants fused to T18, co-expressed in bacteria with human Syt II fused to T25. Based on Clark-Carbon Equation, the number of total colonies needs to be larger than 380 to achieve a 99.8% of possibility of covering all 20 possible amino acids at a single position. Four positions resulted in significant numbers of blue colonies (68 hours after plating). (Figure 5B) Plasmids from blue colonies identified in Figure 5 A were extracted and sequenced to determine the specific residue introduced at the indicated position in BoNT/B-H c . The identified residues are listed. Of note, the blue colonies were further divided to "deep blue colonies", which were found with the position 1191 of

BoNT/B-Hc, and "light blue colonies", which were found with the positions 1183, 1199, and 1178. The term "deep blue colonies" versus "light blue colonies" were artificially judged by the experimenter based on the differences between the blue color of colonies.

[0041] Figure 6A - Figure 6B shows experimental results that indicate the semi-quantitative measurement of the interactions between indicated BoNT/B-Hc mutants with human Syt II in BACTH assay. (Figure 6A) The β-galactosidase activity was measured from lysates of bacteria that express both human Syt II fused with T25 and indicated BoNT/B-H c mutants fused with T18. The activity of β-galactosidase presumably reflects the strength of interactions between human Syt II and BoNT/B-Hc. WT ΒοΝΤ/Β-¾ served as a control. Exchanging residues El 191 to M, C, V, or Q gave the strongest expression of β- galactosidase. Exchanging residues El 191 to S/A/T/N, Yl 183 to C/P, SI 199 to W/E/Y/H, Wl 178 to Y/Q/S showed slight increases of interactions. N = 6 samples/group. (Figure 6B) Binding of indicated BoNT/B-H c mutants to either mouse Syt II (m-Syt II) or human Syt II (h-Syt II), the bindings sites of which are shown in Figure 2A,was assayed by pull down assay. Exchanging residues El 191 to M, C, V, or Q also showed the strongest binding to h- Syt II.

[0042] Figure 7 A - Figure 7B shows experimental results of the screening for secondary mutations in addition to position El 191 that can enhance binding to h-Syt II. (Figure 7 A) Double mutations were created by combining El 191M with all other ten residue changes identified in Figure 5B at positions SI 199/Y1183 AVI 178, respectively. These double mutations were assayed for their ability to bind h-Syt II by pull down assay. Mutants that show strong binding to h-Syt II are indicated with an * (Figure 7B). Proposed mutations for combination that may enhance the affinity between BoNT/B and human Syt II are listed in the table.

[0043] Figure 8 A - Figure 8B show the quantitative measurement of binding affinity between indicated BoNT/B-H c and h-Syt II. (Figure 8A) Binding of GST tagged h-Syt II to His- tagged WT BoNT/B-Hc and El 191M/S1199Y was measured by biolayer interferometry assay. Briefly, anti-GST biosensors were loaded with GST-h-Syt II. The loaded biosensors were incubated with 1 μΜ BoNT/B-H c proteins for measurement of association kinetics, followed by washing steps to measure dissociation kinetics. Representative association and dissociation curves are shown. (Figure 8B) Binding affinities between indicated BoNT/B-Hc and Syt II were measured by biolayer interferometry assay as described in panel A. Kinetic parameters (k on and k off ) and overall binding affinities (K D ) were calculated from a non-linear fit of association and dissociation curves. Binding of WT BoNT/B-Hc to h-Syt II is too weak to be reliably determined, with estimated K D beyond the detection limit (> 20 μΜ), whereas majority of double and triple mutants showed drastically increased binding affinity at the range of 0.59 to 4.30 μΜ.

[0044] Figure 9A- Figure 9C show experimental results that indicate selected BoNT/B-Hc mutants have enhanced binding to human Syt I. (Figure 9A) WT BoNT/B-Hc and El 191M mutant were purified as His6-tagged recombinant proteins and incubated with immobilized GST-tagged h-Syt I (1-80), with or without the presence of co-receptor gangliosides (Gangl). Binding of WT BoNT/B-Hc to h-Syt I requires the presence of the co-receptor gangliosides, indicating a relatively weak interactions between WT BoNT/B-Hc and h-Syt. The El 191M mutant can bind to h-Syt I without gangliosides, showing improved binding affinity to h-Syt I. (Figure 9B) Binding of WT and indicated mutant BoNT/B-H c to h-Syt I were measured by biolayer interferometry as described in Figure 8A. (Figure 9C) Kinetic parameters (k on and k off ) and overall affinities (K D ) for WT BoNT/B-H c , BoNT/B-H c (El 191M/S1199Y) ("B H c MY"), and BoNT/B-H c (El 191 V/Sl 199Y) ("B H c VY") to h-Syt I were measured and calculated as described in Figure 8B. Binding of WT ΒοΝΤ/Β-¾ (B ¾ wt) to h-Syt I cannot be reliably determined, with estimated KD over the detection limit (> 20 μΜ). BoNT/B-H c (El 191M/S1199Y, B H c , MY) and BoNT/B-H c (El 191 V/Sl 199Y, B H c VY) showed drastically increased binding affinity, with K D at 2.9 μΜ and 5.82 μΜ, respectively.

[0045] Figure 10A - Figure 10B are photographs of in situ immunostaining analysis indicating H C B MY showed a robust binding to humanized neurons that express h-Syt II.

(Figure 10A) Humanized neurons were created by knocking down endogenous Syt I and expressing full-length h-Syt II in cultured rat cortical neurons. Neurons that express full- length m-Syt II or m-Syt II (F54L) served as additional controls. These neurons were exposed to WT H C B, followed by immunostaining analysis. Bound H C B was detected via a HA tag fused to H C B. Synapsin was labeled as a marker for presynaptic terminals. WT H C B bound to synapses of WT neurons but not Syt I KD neurons. Expression of full-length m-Syt II via lentiviral transduction restored binding of ¾B, while expression of full-length m-Syt II (F54L) or full-length h-Syt II failed to rescue the binding. (Figure 10B) Syt I KD also abolished binding of H C B MY to neurons. The binding was rescued by expression of full length m-Syt II, m-Syt II (F54L), and h-Syt II.

[0046] Figure 11 A- Figure 11C are experimental results that indicate ΒοΝΤ/Β Μ γ displayed enhanced efficacy on blocking neurotransmission in humanized neurons. (Figure 11 A) Humanized neurons were created as described in Figure 11 A. These neurons were exposed to a gradient of concentrations of full-length WT BoNT/B or ΒοΝΤ/Β Μ γ for 24 hours. Cell lysates were harvested and subjected to immunoblotting analysis, β-tubulin served as an internal loading control. More VAMP2 was cleaved by BoNT/B MY than WT BoNT/B at the same concentrations, indicating that ΒοΝΤ/Β Μ γ entered neurons more efficiently than WT BoNT/B. Humanized neurons were exposed to a gradient of concentrations of WT BoNT/B or BONT/B MY for 24 hours and then the mlPSC activity was recorded by whole-cell patch- clamp approach. (Figure 1 IB) shows representative mlPSC recordings at 30 pM toxins. (Figure 11C) depicts the mlPSC activities versus toxin concentrations, normalized to neurons that were not exposed to any toxins. The half maximum inhibitory concentration (IC50) is 89 pM for WT BoNT/B and 7.8 pM for ΒοΝΤ/Β Μ γ, demonstrating that enhanced binding to human receptors resulted in increased efficacy of toxin at functional levels in neurons. [0047] Figure 12 is the amino acid sequence of the BoNT/B-Hc (strain 1; BoNT/Bl Okra strain). Residues 857-1291 of BoNT/B, strain 1, GenBank: AB232927.1. (SEQ ID NO: 2).

[0048] Figure 13 is the nucleic acid sequence encoding BoNT/B-Hc (strain Bl, Okra strain) residues 857-1291 of BoNT/B, strain 1, based on GenBank: AB232927.1), which has been optimized for expression in E. coli. (SEQ ID NO: 3 )

[0049] Figure 14 shows the amino acid sequence of C. botulinum serotype A (1296 a.a.). (SEQ ID NO: 4)

[0050] Figure 15 shows the amino acid sequence of C. botulinum serotype B (1291 a.a.). (SEQ ID NO: 5)

[0051] Figure 16 shows the amino acid sequence of C. botulinum serotype CI (1291 a.a.). (SEQ ID NO: 6)

[0052] Figure 17 shows the amino acid sequence of C. botulinum serotype D (1276 a.a.). (SEQ ID NO: 7)

[0053] Figure 18 shows the amino acid sequence of C. botulinum serotype E (1252 a.a.). (SEQ ID NO: 8)

[0054] Figure 19 shows the amino acid sequence of C. botulinum serotype F (1274 a.a.). (SEQ ID NO: 9)

[0055] Figure 20 shows the amino acid sequence of C. botulinum serotype G (1297 a.a.). (SEQ ID NO: 10)

[0056] Figure 21 shows the amino acid sequence of C. botulinum serotype B, Strain Eklund 17B (BoNT/B4) (SEQ ID NO: 11) Genbank Ref: EF051570.1.

DETAILED DESCRIPTION OF THE INVENTION

[0057] Aspects of the invention relate to the generation of polypeptides, and nucleic acids encoding the polypeptides, comprising a modified receptor binding domain of Clostridial botulinum (e.g., serotype B (B-H c )), and more particularly to C. botulinum neurotoxin (BoNT) polypeptide which has improved binding to its human receptors through the incorporation of a modified receptor binding domain, and nucleic acids encoding the polypeptide. From these findings, a new generation of therapeutic BoNTs can be created by utilizing the modified receptor binding domain identified herein, with improved efficacy and specificity to target human neurons than the currently utilized WT BoNTs.

Definitions [0058] As used herein, the term "binding affinity" means how strong a molecule's binding activity is for a particular receptor system. In general, high binding affinity results from greater intermolecular force between a binding domain and its receptor system while low binding affinity involves less intermolecular force between the ligand and its receptor. High binding affinity involves a longer residence time for the binding domain at its receptor binding site than is the case for low binding affinity. As such, a molecule with a high binding affinity means a lower concentration of that molecule is required to maximally occupy the binding sites of a receptor system and trigger a physiological response. Conversely, low binding affinity means a relatively high concentration of a molecule is required before the receptor binding sites of a receptor system is maximally occupied and the maximum physiological response is achieved. Thus, a botulinum neurotoxin of the present invention with increased binding activity due to high binding affinity will allow administration of reduced doses of the toxin, thereby reducing or preventing unwanted side-effects associated with toxin dispersal into non-targeted areas.

[0059] One parameter that is often used to measure the binding affinity is defined as binding K D . Because K D is defined as the ratio between dissociation constant (K 0ff ) and association constant (K on ), the lower the K D value, the higher the binding affinity.

[0060] As the term is used herein, "significantly enhanced binding" when used to describe the binding affinity of a C. botulinum neurotoxin molecule or BoNT/B-Hc binding fragment thereof, of the present invention to a specific receptor (e.g., human Syt II or human Syt I), refers to an increase in binding affinity for a specific receptor that is substantially increased (e.g., by 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90% of the binding affinity of the wild type molecule) as compared to the non-substituted version of the molecule. In one embodiment, the enhanced binding of the substituted molecule is demonstrated by a binding affinity that is an order of magnitude or more than the binding affinity of the non-substituted molecule (e.g., the neurotoxin with a naturally occurring BoNT H c molecule). Put another way, the K D of the substituted molecule is an order of magnitude or more lower than the K D of the non- substituted molecule In one embodiment, the enhanced binding is demonstrated by a binding affinity that is significantly higher (e.g., 1.5X, 2. OX, 2.5X, 3. OX, etc.) than the binding affinity of the non-substituted molecule. Put another way, the K D of the substituted molecule is significantly lower (e.g., 1.5X, 2. OX, 2.5X, 3. OX, etc.) than the K D of the non- substituted molecule. In one embodiment, the enhanced binding demonstrated is in the range of about 0.59 μΜ to 5.82 μΜ K D . In one embodiment, the enhanced binding demonstrated is in the range of about 0.59 μΜ to 4.3 μΜ K D . In one embodiment, the enhanced binding is demonstrated by a K D < 5.82 μΜ. In one embodiment, the enhanced binding is demonstrated by a K D < 4.30 μΜ. In one embodiment, the enhanced binding is

demonstrated by a K D ≤ 2.9 μΜ. In one embodiment, the enhanced binding is demonstrated by a K D of about 0.59 μΜ.

[0061] In one embodiment, the K D of the substituted molecule for human Syt II is < 10 μΜ, preferably < 9, 8, 7, 6, 5, 4, 3 or 2 μΜ, more preferably < 1 or 0.6 μΜ. In one embodiment, the K D of the substituted molecule for human Syt I is < 10 μΜ, preferably < 9, 8, 7, 6, 5, 4 μΜ, more preferably < 3 μΜ. In one embodiment, the K D of the substituted molecule for human Syt II is < 7 μΜ and the K D of the substituted molecule for human Syt I is < 6 μΜ. In a preferred embodiment, the K D of the substituted molecule for human Syt II is < 1 μΜ and the K D of the substituted molecule for human Syt I is < 3 μΜ.

[0062] In one embodiment, enhanced binding results in detectable binding to human Syt I in the absence of co-receptor gangliosides, such as exemplified by the experiments described herein.

[0063] The term "significantly enhanced binding" or "significantly reduced binding" when used to describe the binding affinity of a ΒοΝΤ/Β-¾ binding fragment produced by the point mutations described herein refers to an increase or decrease, respectively, in binding affinity of the modified binding domain (e.g., expressed as an isolated fragment or in the context of the larger polypeptide) to a specific receptor (e.g., human Syt II or human Syt I) as discussed directly above.

[0064] As the term is used herein, "significantly reduced binding" when used to describe the binding affinity of botulinum neurotoxin molecule or binding fragment thereof (e.g.,

BoNT/B-Hc), of the present invention to a specific receptor (e.g., human Syt I or human Syt II), refers to a decrease in binding affinity for the specific receptor that is substantially decreased (e.g., by 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90% of the binding affinity of the wild type molecule) as compared to the non- substituted version of the molecule. In one embodiment, the decreased binding of the substituted molecule results in a binding affinity that is an order of magnitude or more less than the binding affinity of the non- substituted neurotoxin (e.g., the neurotoxin with a naturally occurring BoNT H c molecule). Put another way, the K D of the substituted molecule is an order of magnitude or more higher than the K D of the non- substituted neurotoxin. In one embodiment, the reduced binding of the substituted molecule results in a binding affinity that is significantly lower (e.g., 1.5X, 2. OX, 2.5X, 3. OX, etc.) than the binding affinity of the non-substituted molecule. Put another way, the K D of the substituted molecule is significantly higher (e.g., 1.5X, 2. OX, 2.5X, 3. OX, etc.) than the K D of the non- substituted molecule. In one embodiment, significantly reduced binding results in loss of detectable binding to human Syt I in the absence of co-receptor gangliosides.

[0065] As used herein, the term "botulinum neurotoxin" means any polypeptide that can execute the overall cellular mechanism whereby a C. botulinum toxin enters a neuron and inhibits neurotransmitter release. This mechanism encompasses the binding of a C.

botulinum toxin to a low or high affinity receptor complex, the internalization of the toxin, the translocation of the toxin light chain into the cytoplasm and the enzymatic modification of a C. botulinum toxin substrate. The term "polypeptide" and "protein" are used

interchangeably herein, in reference to the C. botulinum neurotoxin molecule and fragments thereof.

[0066] A "modified receptor binding domain" or "modified He", as the term is used herein, facilitates the binding of the C. botulinum neurotoxin molecule in which it is comprised, to a receptor for C. botulinum neurotoxin located on the surface of a target cell. Such a molecule is typically generated through genetic recombination technology. The modified ¾ has a binding activity for the receptor for C. botulinum neurotoxin located on the surface of a target cell. As used herein, the term "binding activity" means that one molecule is directly or indirectly contacting another molecule via at least one intermolecular or intramolecular force, including, without limitation, a covalent bond, an ionic bond, a metallic bond, a hydrogen bond, a hydrophobic interaction, a van der Waals interaction, and the like, or any

combination thereof. "Bound" and "bind" are considered terms for binding.

[0067] As used herein, the term "C. botulinum toxin protease domain" means a C. botulinum toxin domain that can execute the enzymatic target modification step of the intoxication process. Thus, a C. botulinum toxin protease domain specifically targets a C. botulinum toxin substrate and encompasses the proteolytic cleavage of a C. botulinum toxin substrate, such as, e.g., SNARE proteins like a SNAP -25 substrate, a VAMP substrate and a Syntaxin substrate.

[0068] Non-limiting examples of C. botulinum toxin protease domains are provided in Tables 1 and 2.

[0069] As used herein, the term "C. botulinum toxin translocation domain" or "H N " means a C. botulinum toxin domain that can execute the translocation step of the intoxication process that mediates C. botulinum toxin light chain translocation. Thus, a ¾ facilitates the movement of a C. botulinum toxin light chain across a membrane and encompasses the movement of a C. botulinum toxin light chain through the membrane an intracellular vesicle into the cytoplasm of a cell. Non-limiting examples of a H N include a BoNT/A H N , a BoNT/B H N , a BoNT/Cl H N , a BoNT/D H N , a BoNT/E H N , a BoNT/F H N , and a BoNT/G H N , the amino acid sequences of which are provided in Table 1 and Figures 12, 14-20.

[0070] As used herein, the term "C. botulinum receptor-binding domain" is synonymous with "He domain" and means any naturally occurring C. botulinum receptor binding domain that can execute the cell binding step of the intoxication process, including, e.g., the binding of the C. botulinum toxin to a C. botulinum toxin-specific receptor system located on the plasma membrane surface of a target cell. It is envisioned that replacement of the binding activity can be achieved by, e.g., replacing the entire C. botulinum ¾ domain with a modified (e.g., enhanced) H c domain.

[0071] As used herein, the term "C. botulinum toxin target cell" means a cell that is a naturally occurring cell that a naturally occurring C. botulinum toxin is capable of intoxicating, including, without limitation, motor neurons; sensory neurons; autonomic neurons; such as, e.g., sympathetic neurons and parasympathetic neurons; non-peptidergic neurons, such as, e.g., cholinergic neurons, adrenergic neurons, noradrenergic neurons, serotonergic neurons, GABAergic neurons; and peptidergic neurons, such as, e.g., Substance P neurons, Calcitonin Gene Related Peptide neurons, vasoactive intestinal peptide neurons, Neuropeptide Y neurons, cholecystokinin neurons.

[0072] By "isolated" is meant a material that is free to varying degrees from components which normally accompany it as found in its native state. "Isolate" denotes a degree of separation from original source or surroundings, e.g. from flanking DNA or from the natural source of the DNA, or from flanking amino acids.

[0073] The term "purified" is used to refer to a substance such as a polypeptide that is "substantially pure", with respect to other components of a preparation (e.g., other polypeptides). It can refer to a polypeptide that is at least about 50%, 60%, 70%, or 75%, preferably at least about 85%, more preferably at least about 90%, and most preferably at least about 95% pure, with respect to other components. Recast, the terms "substantially pure" or "essentially purified", with regard to a polypeptide, refers to a preparation that contains fewer than about 20%, more preferably fewer than about 15%, 10%, 8%, 7%, most preferably fewer than about 5%, 4%, 3%, 2%, 1%, or less than 1%, of one or more other components (e.g., other polypeptides or cellular components).

[0074] The term "conservative" or "conservative substitution mutation" as used herein refers to a mutation where an amino acid is substituted for another amino acid that has similar properties, such that one skilled in the art of peptide chemistry would expect the secondary structure, chemical properties, and/or hydropathic nature of the polypeptide to be substantially unchanged. The following groups of amino acids have been historically substituted for one another as conservative changes: (1) ala, pro, gly, glu, asp, gin, asn, ser, thr; (2) cys, ser, try, thr; (3) val, ile, leu, met, ala, phe; (4) lys, arg, his; and (5) phe, tyr, trp, his. Other commonly accepted conservative substitutions are listed below:

[0075] The term "substitution mutation" without the reference to a specific amino acid, may include any amino acid other than the wild type residue normally found at that position. Such substitutions may be replacement with non-polar (hydrophobic ) amino acids, such as glycine, alanine, valine, leucine, isoleucine, methionine, phenylalanine, tryptophan, and proline. Substitutions may be replacement with polar (hydrophilic) amino acids such as serine, threonine, cysteine, tyrosine, asparagine, and glutamine. Substitutions may be replacement with electrically charged amino acids e.g,. negatively electrically charged amino acids such as aspartic acid and glutamic acid and positively electrically charged amino acids such as lysine, arginine, and histidine.

[0076] The substitution mutations described herein will typically be replacement with a different naturally occurring amino acid residue, but in some cases non-naturally occurring amino acid residues may also be substituted. Non-natural amino acids, as the term is used herein, are non-proteinogenic (i.e., non-protein coding) amino acids that either occur naturally or are chemically synthesized. Examples include but are not limited to β-amino acids (β3 and β2), homo-amino acids, proline and pyruvic acid derivatives, 3-substituted alanine derivatives, glycine derivatives, ring-substituted phenylalanine and tyrosine derivatives, linear core amino acids, diamino acids, D-amino acids, and N-methyl amino acids. In some embodiments, the amino acid can be substituted or unsubstituted. The substituted amino acid or substituent can be a halogenated aromatic or aliphatic amino acid, a halogenated aliphatic or aromatic modification on the hydrophobic side chain, or an aliphatic or aromatic modification.

[0077] The term "therapeutically effective amount" refers to an amount that is sufficient to effect a therapeutically significant reduction in one or more symptoms of the condition when administered to a typical subject who has the condition. A therapeutically significant reduction in a symptom is, e.g. about 10%, about 20%, about 30%, about 40%, about 50%, about 60%), about 70%, about 80%, about 90%, about 100%, or more as compared to a control or non-treated subject.

[0078] The term "treat" or "treatment" refers to therapeutic treatment wherein the object is to eliminate or lessen symptoms. Beneficial or desired clinical results include, but are not limited to, elimination of symptoms, alleviation of symptoms, diminishment of extent of condition, stabilized (i.e., not worsening) state of condition, delay or slowing of progression of the condition.

[0079] As used herein, a "subject" refers to a human or animal. Usually the animal is a vertebrate such as a primate, rodent, domestic animal or game animal. Primates include chimpanzees, cynomologous monkeys, spider monkeys, and macaques, e.g., Rhesus.

Rodents include mice, rats, woodchucks, ferrets, rabbits and hamsters. Domestic and game animals include cows, horses, pigs, deer, bison, buffalo, feline species, e.g., domestic cat, canine species, e.g., dog, fox, wolf, avian species, e.g., chicken, emu, ostrich, and fish, e.g., trout, catfish and salmon. Patient or subject includes any subset of the foregoing, e.g., all of the above, but excluding one or more groups or species such as humans, primates or rodents. In certain embodiments of the aspects described herein, the subject is a mammal, e.g., a primate, e.g., a human. The terms, "patient" and "subject" are used interchangeably herein. A subject can be male or female. A subject can be a fully developed subject (e.g., an adult) or a subject undergoing the developmental process (e.g., a child, infant or fetus).

[0080] Preferably, the subject is a mammal. The mammal can be a human, non-human primate, mouse, rat, dog, cat, horse, or cow, but are not limited to these examples. Mammals other than humans can be advantageously used as subjects that represent animal models of disorders associated with unwanted neuronal activity. In addition, the methods and compositions described herein can be used to treat domesticated animals and/or pets.

Embodiments [0081] The observation that BoNT/B is less specific and potent in humans due to its inability to bind human Syt II, may explain why comparatively higher doses are required than

BoNT/A. Higher BoNT/B doses correspond to increased chances for triggering antibody responses and for serious side-effects to occur. Therefore, improved binding of BoNT/B to the human receptor Syt II, to increase its efficacy and specificity to target human neurons should allow a reduced amount of the toxin doses used in therapeutic applications.

[0082] Aspects of the invention arise from the finding that modifying the protein sequence of BoNT/B-Hc modifies binding of the fragment containing the receptor binding domain, to the human Syt II receptor. Specific modifications have been identified that enhance binding, thereby generating a domain that binds human Syt II with high-affinity. The modified BoNT/B-Hc, when in the context of a full length BoNT protein, retains these binding properties. Incorporation of a modified receptor binding domain with enhanced binding, into a molecule comprising the other BoNT domains, thereby generates a full length BoNT molecule with similarly enhanced receptor binding. As such, new versions of BoNT with high-affinity binding to human Syt II are generated. BoNT with significantly enhanced binding can be used in similar therapies, albeit at lower doses than presently available BoNT molecules, thus providing safer methods of treatment.

[0083] The BoNT polypeptides, including full-length BoNT polypeptides and BoNT polypeptide fragments or domains described herein (e.g., the modified BoNT/Hc), and nucleic acid molecules which encode them, are explicitly encompassed in the invention. These polypeptides and nucleic acid molecules can be generated by recombinant DNA procedures known in the art. Such polypeptides are typically referred to as "recombinant polypeptides" or "recombinant nucleic acids".

Botulinum Neurotoxin Protein

[0084] One aspect of the invention relates to a botulinum neurotoxin (BoNT) protein comprising a modified receptor binding domain (e.g., of Clostridial botulinum serotype B), as described herein. The BoNT protein further comprises a protease domain, a translocation domain, and a protease cleavage site. Typically these are arranged in a linear amino-to- carboxyl single polypeptide order of the protease domain, the protease cleavage site, the translocation domain and the modified receptor binding domain. However, different arrangements of the various domains are expected to function adequately. In one

embodiment, the modified receptor binding domain comprises one or more substitution mutations which lead to significantly enhanced binding to the human Syt I receptor and/or the human Syt II receptor.

[0085] The BoNT protein may further comprise a domain that is useful for purification of the molecule such as a hexahistidine tag (His6), or an epitope tag such as a hemaglutinin (HA) tag (YPYDVPDYA (SEQ ID NO: 12)). Various such domains are known and used in the art for such purposes.

[0086] BoNT has the overall structure shown in Figure IB. BoNT is comprised of three domains, each domain having a specific and independent function: a protease domain (also referred to as the light chain), a translocation domain (H N ), and a receptor-binding domain (He). Domains of the various strains of C. botulinum neurotoxin have been shown to be largely interchangeable (as demonstrated by naturally occurred chimeric toxins such as BoNT/CD, which is composed of the light chain and H N of BoNT/C, with the H c of BoNT/D 34 , in U.S. Patent 8,052,979, incorporated herein by reference). The protein can be in single chain form or di-chain form. The di-chain form results from the naturally occurring protease processing of a protease cleavage site located between the protease domain and the translocation domain. The protein is maintained in the di-chain form following protease processing by the presence of a di-sulfide bond.

[0087] Strains of Clostridia botulinum produce seven antigenically-distinct types of botulinum toxins, which have been identified by investigating botulism outbreaks in man (BoNT/A, /B, IE and IF), animals (BoNT/Cl and /D), or isolated from soil (BoNT/G). While all seven BoNT serotypes have similar structure and pharmacological properties, each also displays heterogeneous bacteriological characteristics. The genetic diversity of the C.

botulinum strains is described in detail in Hill et al. (Journal of Bacteriology, Vol. 189, No. 3, p. 818-832 (2007)) 35 , the contents of which are incorporated herein by reference. In one embodiment, the BoNT of the invention has domains which are all of the same serotype (A, B, C, D, E, F or G). In one embodiment, one or more of those domains of the same serotype differ as to their strain and/or subtype.

[0088] Toxins from the various C. botulinum serotypes/strains share the same functional domain organization and overall structural architecture. C. botulinum toxins are each translated as a single chain polypeptide of approximately 150 kDa that is subsequently cleaved by proteolytic scission within a disulfide loop by a naturally-occurring protease, such as, e.g., an endogenous C. botulinum toxin protease or a naturally-occurring proteases produced in the environment. This posttranslational processing yields a di-chain molecule comprising an approximately 50 kDa light chain (LC) and an approximately 100 kDa heavy chain (HC) held together by a single disulfide bond and noncovalent interactions. Each mature di-chain molecule comprises three functionally distinct domains: 1) a proteolytic domain located in the LC that includes a metalloprotease region containing a zinc-dependent endopeptidase activity which specifically targets core components of the neurotransmitter release apparatus; 2) a translocation domain contained within the amino-terminal half of the HC (H N ) that facilitates release of the LC from intracellular vesicles into the cytoplasm of the target cell; and 3) a binding domain found within the carboxyl-terminal half of the HC that determines the binding activity and binding specificity of the toxin to the receptor complex located at the surface of the target cell. The locations of the specific domains within the toxin of various serotypes/strains are provided in Table 1 :

TABLE 1

C. botulinum toxin domains from various serotypes/strains

Toxin LC H N He

BoNT/A Ml -K448 A449-K871 N872-L1296

BoNT/B Ml -K441 A442-S858 E859-E1291

BoNT/Cl Ml -K449 T450-N866 N867-E1291

BoNT/D Ml -R445 D446-N862 S863-E1276

BoNT/E Ml -R422 K423-K845 R846-K1252

BoNT/F Ml -K439 A440-K864 K865-E1274

BoNT/G Ml -K446 S447-S863 N864-E1297

[0089] Complete amino acid sequences of the toxins are provided in Figures 14-21.

[0090] The binding, translocation and protease activity of these three functional domains are all necessary for toxicity. The overall cellular intoxication mechanism whereby C. botulinum toxins enter a neuron and inhibit neurotransmitter release is similar, regardless of serotype or subtype. Without wishing to be bound by theory, the intoxication mechanism involves at least four steps: 1) receptor binding, 2) complex internalization, 3) light chain translocation, and 4) protease target modification. The process is initiated when the H c domain of a C. botulinum toxin binds to a toxin-specific receptor located on the plasma membrane surface of a target cell. The binding specificity of a receptor complex is thought to be achieved, in part, by specific combinations of gangliosides and protein receptors. Once bound, the toxin/receptor complexes are internalized by endocytosis and the internalized vesicles are sorted to specific intracellular routes. The translocation step is triggered by the acidification of the vesicle compartment. Once translocated, light chain endopeptidase of the toxin is released from the intracellular vesicle into the cytosol where it specifically targets one of three proteins known as the core components of the neurotransmitter release apparatus (vesicle-associated membrane protein (VAMP)/synaptobrevin, synaptosomal-associated protein of 25 kDa (SNAP -25) and Syntaxin). These core components are necessary for synaptic vesicle docking and fusion at the nerve terminal and constitute members of the soluble N-ethylmaleimide- sensitive factor-attachment protein-receptor (SNARE) family. BoNT/A and BoNT/E cleave SNAP-25 in the carboxyl-terminal region, releasing a nine or twenty-six amino acid segment, respectively, and BoNT/Cl also cleaves SNAP-25 near the carboxyl-terminus. The botulinum serotypes BoNT/B, BoNT/D, BoNT/F and BoNT/G, and tetanus toxin, act on the conserved central portion of VAMP, and release the amino-terminal portion of VAMP into the cytosol. BoNT/Cl cleaves syntaxin at a single site near the cytosolic plasma membrane surface. The selective proteolysis of synaptic SNAREs accounts for the block of neurotransmitter release caused by C. botulinum toxins in vivo. The SNARE protein targets of C. botulinum toxins are common to exocytosis in a variety of non-neuronal types; in these cells, as in neurons, light chain peptidase activity inhibits exocytosis, see, e.g., Yann Humeau et al., How Botulinum and Tetanus Neurotoxins Block Neurotransmitter Release, 82(5) Biochimie. 427-446 (2000); Kathryn Turton et al., Botulinum and Tetanus Neurotoxins: Structure, Function and

Therapeutic Utility, 27(11) Trends Biochem. Sci. 552-558. (2002); Giovanna Lalli et al., The Journey of Tetanus and Botulinum Neurotoxins in Neurons, 11(9) Trends Microbiol. 431- 437, (2003).

Domains and Chimeric Neurotoxins

[0091] The botulinum neurotoxin of the present invention comprises a modified receptor binding domain (H c ). The modified receptor binding domain exhibits significantly enhanced binding to one or more human receptors typically bound and utilized by one or more C.

botulinum toxin strains (e.g., Sytll, Sytl). The modified receptor binding domain can be of any serotype (A, B, C, D, E, F or G), strain or subtype (as described herein). This may be the same or different serotype, strain/subtype as one or more other domains within the BoNT. Examples of specific modified receptor binding domains are provided herein. The isolated modified receptor binding domain polypeptide described herein is also encompassed by the present invention, as is the isolated nucleic acid molecule by which it is encoded. In one embodiment, the H c is serotype B. In one embodiment, the H c is serotype A. [0092] The botulinum neurotoxin of the present invention also comprises a protease domain, also referred to in the art as a light chain variant. The light chain variant may be a naturally occurring light chain variant, such as, e.g., C. botulinum toxin light chain isoforms and C. botulinum toxin light chain subtypes; or a non-naturally occurring C. botulinum toxin light chain variant, such as, e.g., conservative substitution C. botulinum toxin light chain variants. The protease domain can be of any serotype (A, B, C, D, E, F or G), strain or subtype (as described herein). This may be the same or different serotype, strain/subtype as one or more other domains within the BoNT. In one embodiment, the protease domain is serotype B. In one embodiment, the protease domain is serotype A.

[0093] The botulinum neurotoxin of the present invention also comprises a toxin translocation domain (H N ). The toxin translocation domain can be of any serotype (A, B, C, D, E, F or G), strain or subtype (as described herein). This may be the same or different serotype, strain/subtype as one or more other domains within the BoNT. In one embodiment, the H N is serotype B. In one embodiment, the H N is serotype A.

[0094] The various domains described herein (e.g., H N , He, or protease domain) include, without limitation, naturally occurring variants, such as, e.g., isoforms and subtypes; non- naturally occurring variants, such as, e.g., conservative substitution mutations. Non- naturally-occurring variants, refers to a domain that has at least one amino acid change from the corresponding region of the reference sequences (e.g., from Table 1 or Figures 14, 16-23) and can be described in percent identity to the corresponding region of that reference sequence.

[0095] It is recognized by those of skill in the art that within each serotype of C. botulinum toxin there can be naturally occurring C. botulinum domain variants that differ somewhat in their amino acid sequence, and also in the nucleic acids encoding these proteins. A naturally occurring C. botulinum toxin domain (e.g., light chain, H N or ¾) variant envisioned for use in the generation of the BoNT of the present invention can function in substantially the same manner as the reference C. botulinum toxin domain on which the naturally occurring C. botulinum domain variant is based, and can be substituted for the reference C. botulinum toxin domain in any aspect of the present invention.

[0096] A non-limiting example of a naturally occurring C. botulinum toxin domain variant is a C. botulinum toxin domain isoform such as, e.g., a BoNT/A domain isoform, a BoNT/B domain isoform, a BoNT/Cl domain isoform, a BoNT/D domain isoform, a BoNT/E domain isoform, a BoNT/F domain isoform, and a BoNT/G domain isoform. A C. botulinum toxin domain isoform can function in substantially the same manner as the reference C. botulinum toxin domain on which the C. botulinum toxin domain isoform is based, and can be substituted for the reference C. botulinum toxin domain in any aspect of the present invention.

[0097] Another non-limiting example of a naturally occurring C. botulinum toxin domain variant is a C. botulinum toxin domain subtype such as, e.g., a domain from subtype

BoNT/Al, BoNT/A2,BoNT/A3, BoNT/A4, BoNT/A5; a domain from subtype BoNT/Bl, BoNT/B2, BoNT/B3, BoNT/B4, BoNT/B5, B0NT/B6, BoNT/B7; a domain from subtype BoNT/Cl-1, BoNT/Cl-2, BoNT/D-C; a domain from subtype BoNT/El, BoNT/E2,

BoNT/E3, BoNT/E4, BoNT/E5, B0NT/E6, BoNT/E7, B0NT/E8; and a domain from subtype BoNT/Fl, BoNT/F2, BoNT/F3, BoNT/F4, BoNT/F5, B0NT/F6, BoNT/F7. A C. botulinum toxin domain subtype can function in substantially the same manner as the reference C. botulinum toxin domain on which the C. botulinum toxin domain subtype is based, and can be substituted for the reference C. botulinum toxin domain in any aspect of the present invention.

[0098] As used herein, the term "non-naturally occurring variant" (e.g., C. botulinum toxin light chain variant, He and H N ) means a C. botulinum domain produced with the aid of human manipulation, including, without limitation, domains produced by genetic engineering using random mutagenesis or rational design and C. botulinum domains produced by chemical synthesis. Non-limiting examples of non-naturally occurring C. botulinum domain variants include, e.g., conservative C. botulinum domain variants. As used herein, the term "conservative C. botulinum domain variant" means a C. botulinum domain that has at least one amino acid substituted by another amino acid or an amino acid analog that has at least one property similar to that of the original amino acid from the reference C. botulinum domain sequence (e.g., Table 1 and Figures 12, 14-21). The variant may have one, two, three, four, five or more conservative amino acid substitutions compared to the reference domain sequence. Examples of properties include, without limitation, similar size, topography, charge, hydrophobicity, hydrophilicity, lipophilicity, covalent-bonding capacity, hydrogen-bonding capacity, a physicochemical property, of the like, or any combination thereof. A conservative C. botulinum domain variant can function in substantially the same manner as the reference C. botulinum toxin domain on which the conservative C. botulinum toxin domain variant is based, and can be substituted for the reference C. botulinum domain in any aspect of the present invention.

[0099] A non-naturally occurring C. botulinum toxin domain variant may substitute one or more amino acids (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or more) from the reference C. botulinum toxin domain on which the naturally occurring C. botulinum toxin domain is based. A non- naturally occurring C. botulinum toxin domain variant can also possess 95% or more (e.g., 96%, 97%, 98% or 99%) amino acid identity to the reference C. botulinum toxin domain on which the naturally occurring C. botulinum domain variant is based.

[00100] Various non-naturally occurring C. botulinum neurotoxins or specific domains thereof, are described in International Patent Publications W095/32738, W096/33273, WO98/07864 and WO99/17806, each of which is incorporated herein by reference. The C. botulinum neurotoxin or specific domain thereof described herein will typically contain naturally occurring amino acid residues, but in some cases non-naturally occurring amino acid residues may also be present. Therefore, so-called "peptide mimetics" and "peptide analogues", which may include non-amino acid chemical structures that mimic the structure of a particular amino acid or peptide, may also be used within the context of the invention. Such mimetics or analogues are characterized generally as exhibiting similar physical characteristics such as size, charge or hydrophobicity, and the appropriate spatial orientation that is found in their natural peptide counterparts. A specific example of a peptide mimetic compound is a compound in which the amide bond between one or more of the amino acids is replaced by, for example, a carbon-carbon bond or other non-amide bond, as is well known in the art (see, for example Sawyer, in Peptide Based Drug Design, pp. 378-422, ACS, Washington D.C. 1995).

[00101] In one aspect of the invention, the botulinum neurotoxin (BoNT) of the present invention comprises a modified receptor binding domain of C. botulinum serotype B

(BoNT/B-Hc). The modified BoNT/B-H c comprises one or more substitution mutations which lead to significantly enhanced binding to the human Syt I receptor and/or the human Syt II receptor. In one embodiment, the BoNT/B-H c is from BoNT/Bl (GenBank access No. : AB232927.1). The amino acid sequence of BoNT/Bl-H c Okra strain, used as the reference template in the present invention is shown in Figure 12. The generation of B-H c from other strains and subtypes by substitution of the amino acids that correspond to the specified position(s) in Bl described herein is also envisioned, as is the generation of longer molecules that incorporate the B-H c such as complete BoNT or a polypeptide comprising the modified B-H c . Such molecules are also encompassed in the invention. In one embodiment, the BoNT or the polypeptide of the present invention comprises a modified receptor binding domain which is not a BoNT/B4 strain known as the Eklund strain (NCBI Reference Sequence:

YP_001893661.1, Genbank Ref: EF051570.1), shown in Figure 21. In one embodiment, the modified B-Hc is not of strain 3, 7 or 8. [00102] Toxin diffusion and generation of neutralization antibodies is a problem associated with but not limited to BoNT/B, as they are also observed with BoNT/A. Improvement of the binding affinity of BoNT/A to its receptor SV2 would relieve these problems. Because BoNT/B binding to Syt MI has much higher affinity than BoNT/A binding to SV2 14>20>26>27 ; a modified BoNT/B receptor binding domain (BoNT/B-H c ) with the ability to bind human Syt II can also be used to replace BoNT/A-H c to generate a modified chimeric BoNT/A which

28 29 30 may have greater efficacy and specificity for human neurons than WT BoNT/A.

[00103] It is further envisioned that the modified He described above (e.g., BoNT/B-Hc) can be utilized to replace the H c of all other BoNT serotypes/strains. As such, another aspect of the invention is a BoNT polypeptide comprising a modified receptor biding domain (H c ) of serotype (A, B, C, D, E, F or G), joined with one or more domains of a different serotype (A, B, C, D, E, F or G), strain or subtype to thereby produce a chimeric neurotoxin. The He region of each BoNTs is well defined and respective H c regions can be exchanged in the BoNT molecule (e.g., via genetic engineering). Such manipulation is routinely performed by the skilled practitioner via standard PCR fusion of DNA encoding BoNT/B-Hc with the light chain (LC) and translocation domain (H N ), or H N -LC, of other BoNTs, which has been well- established in the art. In addition, the replacement may also be performed using the C- terminal part of BoNT/B-H c (designated as H C c), which is the region containing the binding site for protein receptors and gangliosides in each BoNT. The resulting chimeric toxins will have the ability to target human neurons via binding to human Syt I/II. As a non-limiting example, modified BoNT/B-H c (or BoNT/B-H C c) can be used to replace the H c (or H C c) of BoNT/A. The resulting polypeptides are encompassed by the instant invention. These chimeric toxins may have a higher efficacy and specificity targeting human neurons than WT BoNT/A. Such a chimeric BoNT/A toxin can be used for therapeutic applications in humans and offers significant improvements over WT BoNT/A.

[00104] One aspect of the invention relates to an isolated, purified modified receptor binding domain polypeptide as described herein. In one embodiment, the modified receptor binding domain is ΒοΝΤ/Β-¾ (e.g., from BoNT/Bl). In one embodiment, the B-H c is generated from a different strain and/or subtype of BoNT by substitution of the amino acids that correspond to the specified position(s) in Bl described herein. In one embodiment, the H c is subtype B2, B3, B4, B5, B6 or B7. In one embodiment, the modified receptor binding domain is not from BoNT/B4 (the Eklund strain). In one embodiment, the modified B-Hc is not of strain 3, 7 or 8. [00105] The present invention encompasses mutant full-length BoNT that contains a modified H c (e.g., B-H c ) with amino acid substitutions as described herein, for therapeutic

applications in humans. In one embodiment, the full-length BoNT contains a modified He (e.g., B-Hc) with an amino acid substitution at one or combinations of the amino acid residues corresponding to position El 191, SI 199, Y1183, and Wl 178 of Bl (e.g., selected from those listed in Table 2). In one embodiment, the BoNT contains a modified H c (e.g., B- Hc) with a single amino acid substitution as described herein. In one embodiment, the BoNT contains a modified He (e.g., B-Hc) with two amino acid substitutions as described herein. In one embodiment, the BoNT contains a modified H c (e.g., B-H c ) with three amino acid substitutions as described herein. The mutations can be made in the same manner as disclosed herein for BoNT/Hc, (e.g., using any one of BoNT/B subtypes or any serotype as template). In one embodiment, the mutant full-length BoNT contains a modified Hc that is not from BoNT/B4 (the Eklund strain). In one embodiment, the modified B-Hc is not of strain 3, 7 or 8.

[00106] The resulting BoNT toxin can have significantly enhanced binding to human Syt II, and therefore will achieve higher efficacy and specificity to target human neurons than WT BoNT. The resulting mutant may further maintain similar binding to human Sytl, have significantly enhanced binding to human Sytl, or have significantly reduced binding to human Sytl.

Polypeptides and Chimeric Polypeptides

[00107] Another aspect of the invention relates to a polypeptide comprising the modified receptor binding domain (e.g., of serotype A, B, C, D, E, F or G). In one embodiment, the modified Hc, in the context of the polypeptide, has significantly enhanced binding to human Syt II and/or Syt I, as compared to the analogous polypeptide with WT amino acids at the specific positions modified in the Hc (the wild type counterpart). In one embodiment, the modified receptor binding domain is BoNT/B-Hc (e.g., from BoNT/Bl). In one embodiment, the modified H c is generated from a different serotype, strain and/or subtype of BoNT by substitution of the amino acids that correspond to the specified position(s) in Bl described herein. In one embodiment, the modified Hc is not from BoNT/B4 (the Eklund strain). In one embodiment, the modified B-Hc is not of strain 3, 7 or 8. In one embodiment, the modified receptor binding domain has one or more modifications (amino acid substitutions) in the C-terminal part of BoNT/B-H c (designated as H C c), which is the region containing the binding site for protein receptors and gangliosides in each BoNT. In one embodiment, the resulting polypeptide has the ability to target human neurons via binding to human Sytl/II.

[00108] The polypeptide comprising the modified receptor binding domain may be a fusion protein of the receptor binding domain and another functional polypeptide (e.g., a functional polypeptide utilized in a 2-hybrid system (bait or prey such as T18 or T25, or glutathione S transferase (GST)). This can also be referred to as a chimeric polypeptide molecule.

Alternatively, it may be a fusion of the modified receptor binding domain and a polypeptide tag for identification purposes and/or purification purposes (e.g., a hexahistidine tag (His6), or an epitope tag such as a hemaglutinin (HA) tag (YPYDVPDYA (SEQ ID NO: 12)), or GST), many of which are known and commonly used in the art. The fusion may have a stretch of amino acids between the respective functional domains that facilitates linkage, serves as a cleavage site, or to preserve independent conformation and/or function. In one embodiment, the linkage preserves the function of the modified receptor binding domain. In one embodiment, the linkage masks the function of the modified receptor binding domain. Another aspect of the invention relates to a nucleic acid molecule which encodes any one of such polypeptides.

[00109] The modified BoNT/H c (e.g., B-H c ) can be linked to other agents (e.g., proteins, small molecules, short polypeptides, nucleic acids) covalently (e.g., as a fusion protein) or non-covalently. As such, another aspect of the invention relates to a chimeric molecule comprising a first portion that is a modified receptor binding domain of C.botulinum (e.g., serotype B), as described herein, linked to a second portion. The second portion of the molecule can be a bioactive (or "biologically active") molecule such as a therapeutic agent (e.g., a polypeptide or non-polypeptide drug such as a small molecule or nucleic acid).

Linkage of the first and second portions of the molecule can be covalent (e.g., in the form of a fusion protein) or non-covalent. Methods of such linkage are known in the art and can readily be applied by the skilled practitioner. One such use of a chimeric molecule of the invention is as a delivery tool to target neurons in humans. For example, the modified BoNT/Hc can be linked to other therapeutic agents, to serve as the targeting vehicle to deliver the therapeutic agents to neurons in humans by binding to human Syt I and/or Syt II.

Modifications of the Receptor Binding Domain (He)

[00110] As discussed herein, the invention relates to a modified He and polypeptides comprising the modified He (e.g., BoNT or a fusion or chimeric polypeptide). The modification of the H c amino acid sequence used to generate these various polypeptides of the invention can be performed by various methods known to the skilled practitioner.

Examples include, without limitation, targeted mutagenesis (site-directed mutagenesis) or random mutagenesis of each amino acid residue within the region known for binding Syt VII. These Syt binding regions are well defined by previous studies relating to mouse or rat Syt

1 29 36 31 32

receptors ' ' ' ' but have not been clearly determined for interactions between BoNT/B-H c and human Syt receptors. Different subtypes of BoNT/B, or other serotypes which bind to Syt I/II (D-C or G), can be used as the template to create the same or similar mutations by generating corresponding mutations described herein for B 1-¾. The corresponding position for selected residues to be mutated can be readily identified by sequence alignment with the Bl subtype. The resulting polypeptide products are encompasses by the instant invention, as are polypeptides comprising said products and nucleic acid molecules encoding said polypeptides and products.

[00111] Amino acid sequence modifications of H c to produce the modified receptor binding domain can be mutation of a single residue to a different amino acid (single site substitution), mutation of multiple residues at the same time (multiple sites substitution), deletion of one or more residues (deletion), and insertion of one or more residues (insertion), as well as combinations thereof. Methods for mutating proteins are well-known in the art (e.g., targeted single site and multiple sites substitutions on the DNA encoding the BoNT/H c sequence).

[00112] In one embodiment, one or more residues in He that either contact rodent Syt II or the surrounding regions, based on previous literatures on BoNT/B receptor binding domain 29 and reported BoNT/B-Syt II structure (PDB ID: 2 M1) 31 ' 32 , are modified. These include, without limitation those positions that correspond to position Yl 181, PI 197, Al 196, F1204, F1194, P1117, W1178, Y1183, V1118, S1116, K1113, K1192, S1199, S1201, E1191, E1245, Y1256, Dl 115, E1203 of BoNT/B-Bl . In one embodiment, one or more of these residues is systemically replaced with other amino acids. Combinations of various modifications are also envisioned, including, without limitation, mutations of two or more recited positions, to any variety of amino acids, such as those recited herein.

[00113] Multiple site substitutions (e.g., 2 or 3) can be generated by combining mutations in these identified key residues. Such multiple site substitution mutants may have even further enhanced binding to h-Syt II and may further maintain or have enhanced binding to human Syt I. In one embodiment, the modified ¾ has a first amino acid substitution that corresponds to El 191 of serotype B, strain 1, substituted with either Q, M, C, V, L, or Y. In one embodiment, the modified H c has a second site substitution that corresponds to either S1199W, S1199E, S1199H, S1199Y, W1178Y, W1178Q, W1178A, W1178S, Y1183C, Yl 183P, SI 199F or SI 199L of serotype B, strain 1. In one embodiment, the modified ¾ has a multiple site substitution that corresponds to either El 191M and SI 199W, El 191M and W1178Q, E1191C and S1199W, E1191C and S1199Y, E1191C and W1178Q, E1191Q and SI 199W, El 191 V and SI 199W, El 191 V and SI 199Y, or El 191 V and Wl 178Q, of serotype

B, strain 1.

[00114] In one embodiment, the modified receptor binding domain comprises a substitution mutation at a position corresponding to Yl 181 , P 1 197, Al 196, F 1204, F 1194, P 1117, W1178, Y1183, V1118, SI 116, K1113, K1192, S1199, S1201, E1191, E1245, D1115, E1203 or Y1256 of serotype B, strain 1. In one embodiment, the substitution mutation is A, R, N, D, C, Q, E, G, H, I, L, K, M, F, P, T, W, Y or V substituted for S. In one embodiment, the position corresponds to SI 199 or SI 201 of serotype B, strain 1. In one embodiment, the substitution mutation is W, E or H. In one embodiment, the substitution mutation is at SI 199, and is W, E or H. In one embodiment, the substitution mutation is at S1201 and is V. In one embodiment, the positions corresponds to Wl 178, Yl 183, or El 191. In one embodiment, the position corresponds to Wl 178 and the substitution mutation is Y, Q, A or S. In one embodiment, the position correspond to Yl 183 and the substitution mutation is C or P. In one embodiment, the position corresponds to El 191 and the substitution mutation is

C, V, L, or Y.

[00115] A modified He with a single amino acid substitution mutation shown below in Table 2 is also environed, as is its incorporation in the various polypeptides described herein.

Table 2

[00116] In one embodiment, the He comprises one or more mutations that correspond to El 191C/V/L/Y (El 191C, El 191 V, El 191L, or El 191 Y), SI 199W/E/H (SI 199W, SI 199E, or SI 199H), Wl 178Y/Q/A/S (Wl 178Y, Wl 178Q, Wl 178 A, or Wl 178S), and/or Yl 183C/P (Yl 183C or Yl 183P) of serotype B, strain 1 (Figure 3 A, B).

[00117] In one embodiment, the He has two substitution mutations wherein one is a substitution mutation corresponding to position 1191 selected from Table 2. In one embodiment, the He has enhanced binding to h-Syt II compared to the analogous polypeptide having WT amino acids at the specified positions (also referred to herein as the wild type counterpart). In one embodiment, the generated mutant also maintains or has significantly enhanced binding to human Syt I as compared to the wild type counterpart. In particular, the mutation is El 191C, El 191V, El 191L, or El 191 Y. More particularly, the mutation that corresponds to position El 191C, El 191 V, El 191L, or El 191 Y is combined with a second mutation (e.g., such as those described herein) to generate a modified ¾ that has enhanced binding to h-Syt II compared to the analogous polypeptide having WT amino acids at the specified positions (also referred to herein as the wild type counterpart). In one embodiment, the generated mutant also maintains or has significantly enhanced binding to human Syt I as compared to the wild type counterpart (Figure 4A). In one embodiment, the second mutation is a substitution mutation corresponding to a substitution shown for position 1183, 1199, 1201, 1178, 1117, or 1181 in Table 2. In one embodiment, the second mutation is SI 199W, S1199E, S1199H, SI 199Y, Wl 178Y, Wl 178Q, Wl 178A, Wl 178S, Y1183C, Y1183P, SI 199F or SI 199L. In one embodiment the ¾ has two substitution mutations that correspond to those shown for serotype B, strain 1, in Table 3 :

Table 3

E1191M and S1199F E1191Q and S1199F E1191M and W1178Q

E1191Q and W1178Q

E1191C and W1178Q

E1191V and W1178Q

[00118] In one embodiment, the H c has three substitution mutations, also referred to herein as a triple mutation. In one embodiment of the triple mutation, the first mutation corresponds to

E1191Q, E1191M, E1191C, E1191V, E1191L, E1191Y, S1199W, S1199E, S1199H,

SI 199Y, SI 199F, SI 199L, Yl 183C, Yl 183P, Wl 178Y, Wl 178Q, Wl 178A or Wl 178S of serotype B, strain 1. In one embodiment, the second mutation corresponds to El 191Q,

E1191M, E1191C, El 191V, E1191L, El 191 Y, S1199W, S1199E, S1199H, S1199Y,

SI 199F, SI 199L, Yl 183C, Yl 183P, Wl 178Y, Wl 178Q, Wl 178A or Wl 178S of serotype

B, strain 1 (exclusive of the position serving as the first mutation). In one embodiment, the third substitution corresponds to El 191Q, El 191M, El 191C, El 191 V, El 191L, El 191 Y,

S1199W, S1199E, S1199H, S1199Y, S1199F, SI 199L, Yl 183C, Yl 183P, Wl 178Y,

Wl 178Q, Wl 178 A or Wl 178S of serotype B, strain 1 (exclusive of the positions serving as the first and second mutations). In one embodiment of the invention, the double or triple mutation does not contain a Q substituted at both positions corresponding to El 191 and

W1178.

[00119] In one embodiment of the triple mutation, one mutation is a substitution mutation that corresponds to a substitution shown for El 191 in Table 2 (e.g., El 191Q/M/C/V/L or Y). The second mutation may be a substitution mutation corresponding to a substitution shown for SI 199 in Table 2 (e.g., SI 199W/E/H/Y/F or L). The third mutation may be a substitution mutation corresponding to a substitution shown for Wl 178 in Table 2 (e.g., Wl 178Y/Q/A or S). Alternatively, the third mutation may be a substitution mutation corresponding to a substitution shown for Yl 183 in Table 2. (e.g., Yl 183C, Yl 183P). In one embodiment, the triple mutation corresponds to El 191M/S1199W/W1178Q.

Nucleic Acid Molecules

[00120] Another aspect of the invention relates to an isolated nucleic acid molecule comprising a nucleotide sequence that encodes the polypeptides described herein (e.g., modified receptor binding domain, or a polypeptide comprising the modified receptor binding domain, or the botulinum neurotoxin comprising the modified receptor binding domain, described herein). In one embodiment, the nucleic acid molecule comprises the nucleic acid sequence shown in Figure 13. Such nucleic acid molecules can be produced by the skilled practitioner, for example by recombinant DNA techniques. The desired amino acid substitution mutation is made, for example, by modification of the encoding DNA. For example, nucleic acid sequences coding for the polypeptides described herein are generated by mutating the nucleic acid codon encoding the specified amino acid to the desired amino acid using the genetic code shown below:

[00121] In one embodiment, the nucleic acid sequence is optimized for expression in E.coli (e.g., the nucleic acid sequence based on GenBank AB232927.1, the relevant portion of which is shown in Figure 13).

[00122] Another aspect of the invention relates to a nucleic acid vector comprising the nucleic acid molecule described herein. In one embodiment the vector is an expression vector. Such an expression vector is referred to herein as an expression construct, and comprises a nucleic acid molecule disclosed herein operably-linked to the expression vector useful for expressing the nucleic acid molecule in a cell or cell-free extract. A wide variety of expression vectors can be employed for expressing a nucleic acid molecule encoding a C. botulinum neurotoxin of the present invention including, without limitation, a viral expression vector; a prokaryotic expression vector; eukaryotic expression vectors, such as, e.g., a yeast expression vector, an insect expression vector and a mammalian expression vector; and a cell-free extract expression vector. It is further understood that expression vectors useful to practice aspects of these methods may include those which express the C. botulinum neurotoxin under control of a constitutive, tissue-specific, cell-specific or inducible promoter element, enhancer element or both. Non-limiting examples of expression vectors, along with well-established reagents and conditions for making and using an expression construct from such expression vectors are readily available from commercial vendors that include, without limitation, BD

Biosciences-Clontech, Palo Alto, Calif ; BD Biosciences Pharmingen, San Diego, Calif ; Invitrogen, Inc, Carlsbad, Calif; EMD Biosciences-Novagen, Madison, Wis.; QIAGEN, Inc., Valencia, Calif; and Stratagene, La Jolla, Calif. The selection, making and use of an appropriate expression vector are routine procedures well within the scope of one skilled in the art and from the teachings herein.

Cells

[00123] Another aspect of the invention relates to a cell comprising the nucleic acid molecule or expression construct described herein. The cell can be for propagation of the nucleic acid or for expression of the nucleic acid, or both. Such cells include, without limitation, prokaryotic cells including, without limitation, strains of aerobic, microaerophilic, capnophilic, facultative, anaerobic, gram-negative and gram-positive bacterial cells such as those derived from, e.g., Escherichia coli, Bacillus subtilis, Bacillus licheniformis,

Bacteroides fragilis, Clostridia perfringens, Clostridia difficile, Caulobacter crescentus, Lactococcus lactis, Methyl obacterium extorquens, Neisseria meningirulls, Neisseria meningitidis, Pseudomonas fluorescens and Salmonella typhimurium; and eukaryotic cells including, without limitation, yeast strains, such as, e.g., those derived from Pichia pastoris, Pichia methanolica, Pichia angusta, Schizosaccharomyces pombe, Saccharomyces cerevisiae and Yarrowia lipolytica; insect cells and cell lines derived from insects, such as, e.g., those derived from Spodoptera frugiperda, Trichoplusia ni, Drosophila melanogaster and Manduca sexta; and mammalian cells and cell lines derived from mammalian cells, such as, e.g., those derived from mouse, rat, hamster, porcine, bovine, equine, primate and human. Cell lines may be obtained from the American Type Culture Collection, European Collection of Cell Cultures and the German Collection of Microorganisms and Cell Cultures. Non-limiting examples of specific protocols for selecting, making and using an appropriate cell line are described in e.g., INSECT CELL CULTURE ENGINEERING (Mattheus F. A. Goosen et al. eds., Marcel Dekker, 1993); INSECT CELL CULTURES: FUNDAMENTAL AND

APPLIED ASPECTS (J. M. Vlak et al. eds., Kluwer Academic Publishers, 1996); Maureen A. Harrison & Ian F. Rae, GENERAL TECHNIQUES OF CELL CULTURE (Cambridge University Press, 1997); CELL AND TISSUE CULTURE: LABORATORY PROCEDURES (Alan Doyle et al eds., John Wiley and Sons, 1998); R. Ian Freshney, CULTURE OF

ANFMAL CELLS: A MANUAL OF BASIC TECHNIQUE (Wiley-Liss, 4 sup.th ed. 2000); ANFMAL CELL CULTURE: A PRACTICAL APPROACH (John R. W. Masters ed., Oxford University Press, 3 sup.rd ed. 2000); MOLECULAR CLONF G A LABORATORY

MANUAL, supra, (2001); BASIC CELL CULTURE: A PRACTICAL APPROACH (John M. Davis, Oxford Press, 2 sup.nd ed. 2002); and CURRENT PROTOCOLS FN

MOLECULAR BIOLOGY, supra, (2004). These protocols are routine procedures within the scope of one skilled in the art and from the teaching herein.

[00124] The cell can be for expression of the nucleic acid to thereby generate the encoded polypeptide. As such, another aspect of the invention is a method for producing a botulinum neurotoxin protein, an isolated HC, or a polypeptide comprising a modified HC, described herein. Such polypeptides are produced by culturing the host cell that has within it a nucleic acid that encodes the polypeptide, in the context of an expression construct. Culturing is performed under condition suitable for production of the BoNT polypeptide. The expressed polypeptide can be recovered from the culture, purified and formulated by methods known in the art. The expressed polypeptide can also be activated as necessary by methods known in the art. One method of activating the expressed polypeptide is through cleavage (or nicking) by proteases into an active dichain form. Such methods can be adapted from those known in the art, for example as disclosed by Peter F. Bonventre and Lloyd L. KLempe( J. Bacterid 1960, 79(1): 23 and Michaeal L. Dekleva and Bibhuti R. DasGupta (Biochemical and Biophysical Research Communicaitons 1989, 162: 767-772).

Pharmaceutical Compositions

[00125] Another aspect of the present invention relates to a pharmaceutical composition comprising the C. botulinum neurotoxin, or chimeric molecule described herein. In one embodiment, the polypeptide described herein is an active ingredient in a composition comprising a pharmaceutically acceptable carrier (referred to herein as a pharmaceutical composition). A "pharmaceutically acceptable carrier" means any pharmaceutically acceptable means to mix and/or deliver the targeted delivery composition to a subject. The term "pharmaceutically acceptable carrier" as used herein means a pharmaceutically acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting the subject agents from one organ, or portion of the body, to another organ, or portion of the body. Each carrier must be "acceptable" in the sense of being compatible with the other ingredients of the

composition and is compatible with administration to a subject, for example a human. Such compositions can be specifically formulated for administration via one or more of a number of routes, such as the routes of administration described herein. Supplementary active ingredients also can be incorporated into the compositions. When an agent, formulation or pharmaceutical composition described herein, is administered to a subject, preferably, a therapeutically effective amount is administered. As used herein, the term "therapeutically effective amount" refers to an amount that results in an improvement or remediation of the condition. In one embodiment, the pharmaceutical composition is formulated for

administration by injection. In one embodiment, the pharmaceutical composition involves the botulinum neurotoxin encapsulated in microspheres. In one embodiment, the

pharmaceutical composition involves the botulinum neurotoxin formulated for transepithelial delivery. In one embodiment, the pharmaceutical composition involves the botulinum neurotoxin formulated for slow release.

[00126] In one embodiment, the botulinum neurotoxin, polypeptide, or chimeric molecule of the present invention is in the form of a controlled release formula. Such compositions and methods for administration are provides in U.S. Patent publication No. 2007/0020295, the contents of which are herein incorporated by reference.

[00127] Botulinum neurotoxin can be obtained by establishing and growing cultures of Clostridium botulinum in a fermenter and then harvesting and purifying the fermented mixture in accordance with known procedures. All the botulinum toxin serotypes are initially synthesized as inactive single chain proteins which must be cleaved or nicked by proteases to become neuroactive. The bacterial strains that make botulinum toxin serotypes A and G possess endogenous proteases and serotypes A and G can therefore be recovered from bacterial cultures in predominantly their active form. In contrast, botulinum toxin serotypes Ci, D and E are synthesized by nonproteolytic strains and are therefore typically unactivated when recovered from culture. Serotypes B and F are produced by both proteolytic and nonproteolytic strains and therefore can be recovered in either the active or inactive form. The proteolytic strains that produce, for example, the botulinum toxin type B serotype may only cleave a portion of the toxin produced. The exact proportion of nicked to unnicked molecules depends on the length of incubation and the temperature of the culture. Therefore, a certain percentage of a preparation of, for example, the botulinum toxin type B toxin may be inactive. In one embodiment, the neurotoxin of the present invention is in an active state. In one embodiment, the neurotoxin is in an inactive state. In one embodiment, a combination of active and inactive neurotoxin is envisioned.

Kits

[00128] Also encompassed in the present invention is a kit comprising the BoNT or polypeptide disclosed herein (e.g., in the form of a pharmaceutical composition). The kit may comprise one or more of the compositions described herein packaged in a vial. The kit may further comprise a delivery tool or device for the therapeutic administration of the composition, and/or instructions for therapeutic administration. The kit may have all components therein packaged into a formed container.

[00129] Another aspect of the invention relates to a delivery tool or device for administration of the pharmaceutical compositions described herein, pre-loaded with the pharmaceutical composition (e.g., for single use). Such devices may be a syringe or a microneedle device for delivery of the compositions. The syringe may be a single use syringe pre-loaded with an effective amount of the composition. The microneedle device may comprise one or more microneedles coated with the composition described herein, such as is described in U.S. Patent Publication 2010/0196445, the contents of which are incorporated herein in their entirety.

Methods of Treatment

[00130] The present invention also includes methods for treating a condition typically treated with a neurotoxin (e.g, skeletal muscle conditions, smooth muscle conditions, glandular conditions, a neuromuscular disorder, an autonomic disorder, pain, or an aesthetic/cosmetic condition). Such conditions are associated with unwanted neuronal activity, as determined by the skilled practitioner. The method comprises the step of administering a therapeutically effective amount of a BoNT or polypeptide/chimeric molecule described herein (e.g. as a pharmaceutical composition) to the appropriate location in the mammal to reduce the unwanted neuronal activity, to thereby treat the condition. Administration is by a route that contacts an effective amount of the composition to neurons exhibiting the unwanted activity. [00131] Specific conditions envisioned for treatment by the methods discussed herein include, without limitation, spasmodic dysphonia, spasmodic torticollis, laryngeal dystonia, oromandibular dysphonia, lingual dystonia, cervical dystonia, focal hand dystonia, blepharospasm, strabismus, hemifacial spasm, eyelid disorder, cerebral palsy, focal spasticity and other voice disorders, spasmodic colitis, neurogenic bladder (i.e. all diseases involving urinary incontinence, such as e.g., neurogenic detrusor overactivity or idiopathic overactive bladder), prostate cancer and other cancer forms, anismus, limb spasticity, tics, tremors, bruxism, anal fissure, achalasia, dysphagia and other muscle tone disorders and other disorders characterized by involuntary movements of muscle groups, lacrimation,

hyperhydrosis, excessive salivation, excessive gastrointestinal secretions as well as other secretory disorders, pain from muscle spasms, neuropathic pain, inflammatory pain, heachache pain, such as e.g. migraine, itch (pruritis), acne. In addition, the present invention can be used to treat dermatological or aesthetic/cosmetic conditions, for example, reduction of brow furrows, reduction of skin wrinkles. The present invention can also be used in the treatment of sports injuries.

[00132] In addition, the modified neurotoxin can be administered to treat other neuromuscular disorders using well known techniques that are commonly performed with botulinum type A. For example, the present invention can be used to treat pain, for example, headache pain, pain from muscle spasms and various forms of inflammatory pain. For example, Aoki U.S. Pat. No. 5,721,215 and Aoki U.S. Pat. No. 6,113,915 disclose methods of using botulinum toxin type A for treating pain. The disclosure of these two patents is incorporated in its entirety herein by reference.

[00133] Autonomic nervous system disorders can also be treated with a modified neurotoxin. For example, glandular malfunctioning is an autonomic nervous system disorder. Glandular malfunctioning includes excessive sweating and excessive salivation. Respiratory

malfunctioning is another example of an autonomic nervous system disorder. Respiratory malfunctioning includes chronic obstructive pulmonary disease and asthma. Sanders et al. in U.S. Patent No. 5,766,605 disclose methods for treating the autonomic nervous system; for example, treating autonomic nervous system disorders such as excessive sweating, excessive salivation, asthma, etc., using naturally existing botulinum toxins. The disclosure of Sander et al. is incorporated in its entirety by reference herein. In one embodiment, substantially similar methods to that of Sanders et al. can be employed, but using a modified neurotoxin, to treat autonomic nervous system disorders such as the ones discussed above. For example, a modified neurotoxin can be locally applied to the nasal cavity of the mammal in an amount sufficient to degenerate cholinergic neurons of the autonomic nervous system that control the mucous secretion in the nasal cavity.

[00134] Pain that can be treated by a modified neurotoxin includes pain caused by muscle tension, or spasm, or pain that is not associated with muscle spasm. For example, Binder in U.S. Pat. No. 5,714,468 discloses that headache caused by vascular disturbances, muscular tension, neuralgia and neuropathy can be treated with a naturally occurring botulinum toxin, for example Botulinum type A. The disclosures of Binder are incorporated in its entirety herein by reference. In one embodiment, substantially similar methods to that of Binder can be employed, but using a modified neurotoxin, to treat headache, especially the ones caused by vascular disturbances, muscular tension, neuralgia and neuropathy. Pain caused by muscle spasm can also be treated by an administration of a modified neurotoxin. For example, WO2006001676 (Kang Ahn) discloses methods of using a botulinum neurotoxin for treating knee joint pain caused by saphenous nerve entrapment, , WO2008059126 (Christine Favre et al.) discloses methods of using a botulinum neurotoxin for treating pain induced by chemotherapy, WO2008090287 (Christine Favre et al.) discloses methods of using a botulinum neurotoxin for treating pain induced by anti-HIV treatment, WO2009130600 (Christine Favre et al.) discloses methods of using a botulinum neurotoxin for treating pain associated with diabetic neuropathy, and WO2007144493 (Michel Auguet et al.) discloses methods of using a botulinum neurotoxin in combination with an opiate derivative for treating pain. The disclosures of WO2006001676, WO2008059126, WO2008090287, WO2009130600, WO2007144493 are incorporated in its entirety herein by reference.

Furthermore, a modified neurotoxin can be administered to a mammal to treat pain that is not associated with a muscular disorder, such as spasm. In one broad embodiment, methods of the present invention to treat non-spasm related pain include central administration or peripheral administration of the modified neurotoxin.

[00135] An acute or chronic pain that is not associated with a muscle spasm can also be alleviated with a local, peripheral administration of the modified neurotoxin to an actual or a perceived pain location on the mammal. In one embodiment, the modified neurotoxin is administered subcutaneously at or near the location of pain, for example, at or near a cut. In some embodiments, the modified neurotoxin is administered intramuscularly at or near the location of pain, for example, at or near a bruise location on the mammal. In some embodiments, the modified neurotoxin is injected directly into a joint of a mammal, for treating or alleviating pain caused by arthritic conditions. Also, frequent repeated injection or infusion of the modified neurotoxin to a peripheral pain location is within the scope of the present invention

[00136] Routes of administration for such methods are known in the art and easily adapted to the methods described herein by the skilled practitioner (e.g., see for example, Harrison's Principles of Internal Medicine (1998), edited by Anthony Fauci et al., 14th edition, published by McGraw Hill). By way of non-limiting example, the treatment of a

neuromuscular disorder can comprise a step of locally administering an effective amount of the molecule to a muscle or a group of muscles, the treatment of an autonomic disorder can comprise a step of locally administering an effective of the molecule to a gland or glands, and the treatment of pain can comprise a step of administering an effective amount of the molecule the site of the pain. In addition, the treatment of pain can comprise a step of administering an effective amount of a modified neurotoxin to the spinal cord.

[00137] It is also envisioned that the modified H c (e.g., B-H c ) described herein can be utilized as a delivery tool to target neurons and other cell types that express human synaptotagmin II in humans. For example, the modified He linked to another bioactive molecule (e.g., therapeutic agent) covalently or non-covalently to thereby form a chimeric molecule described herein, can serve as the targeting vehicle to deliver the bioactive molecule to neurons and other cell types that express human synaptotagmin II in humans by binding to human Syt I and/or Syt II. As such, another aspect of the invention relates to the use of a chimeric polypeptide molecule described herein to deliver the bioactive molecule to a neuron in a human subject. The second portion of the molecule can be a bioactive molecule such as a therapeutic agent (e.g., a polypeptide or drug). Linkage of the first and second portions of the molecule can be covalent (e.g., in the form of a fusion protein) or non-covalent. Methods of such linkage are known in the art and can readily be applied by the skilled practitioner. The chimeric polypeptide molecule would be administered by a route that resulted in contacting of the polypeptide to the neurons expressing the receptor to which the modified B- Hc specifically binds (the target neuron), as described herein.

Identifying Receptor Binding Activity

[00138] Another aspect of the invention relates to a method for identifying a modified BoNT receptor binding domain for its ability to bind to a receptor (e.g. a human Syt I or human Syt II, or human SV2). The method utilizes the 2-hybrid assay system and utilizes fusions proteins made of the receptor and of the modified Hc respectively fused (expressed as fusions proteins) to the respective "bait" and "prey" subunits used in the 2-hybrid assay (e.g., Gal 4 transcription activation system utilizing an activation domain and a DNA binding domain). The 2-hybrid assay is typically performed in yeast (S. cerevisiae), but similar assay systems have been developed for use in other single celled organisms as well such as in E. coli.

Those systems are equally comparable. In one embodiment, the bacterial adenylate cyclase 2-hybrid assay is used (Karimova et al., Proc Natl Acad Sci U S A. May 12, 1998; 95(10): 5752-5756, the contents of which are incorporated herein by reference). The system uses T18 and T25 as bait and prey, and can utilize E. coli BTH101 cells.

[00139] The modified He is expressed as a fusion protein with T18 (referred to as a first fusion protein) in the E. coli of the 2-hybrid assay. In the method, the receptor (or a binding fragment thereof such as h-Syt II a.a. 1-87) is co-expressed as a fusion protein with T25 (referred to as a second fusion protein) in the E. coli of the 2-hybrid assay. The assay utilizes a positive indicator of interaction between the respective molecules via their respective fusions. One possible positive indicator is color development. Clonal E. coli colonies expressing both the first and second fusion proteins are grown on solid media containing the appropriate selective and reporting media (e.g., ampicillin, kanamycin, X-gal, and IPTG) for an amount of time to allow for generation of a positive indication (e.g., color generation) from positive colonies. Binding of the modified binding domain to the receptor fragment will lead to the positive indication (e.g., expression of LacZ gene and result in generation of a blue color in colonies grown on X-gal plates).

[00140] The colonies are analyzed for such positive indication (e.g., color development) when compared to appropriate controls (positive and negative). The analyzing can be visually or by a non-human machine. An appropriate negative control that fails to produce appreciable positive indication (e.g., LacZ expression) is used (e.g, a clonal colony lacking a key component of the assay system such as having bait and prey that are expressed without a fusion). An appropriate strong positive control can also be used. One example of a strong positive control for the human Syt II receptor in the assay is a Bl-Hc with a substitution mutation at El 191 (M/C/V or Q). A weakly positive control can also be used to identify the strength of the binding. One example of a weakly positive control for human Syt II receptor in the assay is a Bl-Hc with a substitution mutation at Wl 178 (Q/Y/A or S). Identification of the positive indication (e.g., color development) indicates that the modified He binds the receptor. Identification of strong positive indication such as strong color development (e.g. above a weakly positive control) indicates the He binds the receptor with high affinity.

[00141] The modified He used in the assay can be any modified He disclosed herein. By way of non-limiting example, the modified He may contain one, two or three substitution mutations such as those disclosed herein. The modified He may be of any serotype/strain or subtype as disclosed herein.

[00142] The embodiments described here and in the following examples are for illustrative purposes only, and various modifications or changes apparent to those skilled in the art are included within the scope of the invention.

[00143] Unless otherwise defined herein, scientific and technical terms used in connection with the present application shall have the meanings that are commonly understood by those of ordinary skill in the art. Further, unless otherwise required by context, singular terms shall include pluralities and plural terms shall include the singular.

[00144] It should be understood that this invention is not limited to the particular

methodology, protocols, and reagents, etc., described herein and as such may vary. The terminology used herein is for the purpose of describing particular embodiments only, and is not intended to limit the scope of the present invention, which is defined solely by the claims.

[00145] Other than in the operating examples, or where otherwise indicated, all numbers expressing quantities of ingredients or reaction conditions used herein should be understood as modified in all instances by the term "about." The term "about" when used to describe the present invention, in connection with percentages means ±1%.

[00146] In one respect, the present invention relates to the herein described compositions, methods, and respective component(s) thereof, as essential to the invention, yet open to the inclusion of unspecified elements, essential or not ("comprising"). In some embodiments, other elements to be included in the description of the composition, method or respective component thereof are limited to those that do not materially affect the basic and novel characteristic(s) of the invention ("consisting essentially of). This applies equally to steps within a described method as well as compositions and components therein. In other embodiments, the inventions, compositions, methods, and respective components thereof, described herein are intended to be exclusive of any element not deemed an essential element to the component, composition or method ("consisting of).

[00147] All patents, patent applications, and publications identified are expressly incorporated herein by reference for the purpose of describing and disclosing, for example, the

methodologies described in such publications that might be used in connection with the present invention. These publications are provided solely for their disclosure prior to the filing date of the present application. Nothing in this regard should be construed as an admission that the inventors are not entitled to antedate such disclosure by virtue of prior invention or for any other reason. All statements as to the date or representation as to the contents of these documents is based on the information available to the applicants and does not constitute any admission as to the correctness of the dates or contents of these documents.

References for Background and Detailed Description

1. Schiavo, G., Matteoli, M. & Montecucco, C. Neurotoxins affecting neuroexocytosis. Physiol Rev 80, 717-766 (2000).

2. Johnson, E.A. Clostridial toxins as therapeutic agents: benefits of nature's most toxic proteins. Annu Rev Microbiol 53, 551-575 (1999).

3. Aoki, K.R. Botulinum toxin: a successful therapeutic protein. Curr Med Chem 11, 3085-3092 (2004).

4. Montecucco, C. & Molgo, J. Botulinal neurotoxins: revival of an old killer. Curr Opin Pharmacol 5, 274-279 (2005).

5. Lange, O., et al. Neutralizing antibodies and secondary therapy failure after treatment with botulinum toxin type A: much ado about nothing? Clin Neuropharmacol 32, 213-218 (2009).

6. Chapman, M.A., Barron, R., Tanis, D.C., Gill, C.E. & Charles, P.D. Comparison of botulinum neurotoxin preparations for the treatment of cervical dystonia. Clin Ther 29, 1325- 1337 (2007).

7. Cote, T.R., Mohan, A.K., Polder, J.A., Walton, M.K. & Braun, M.M. Botulinum toxin type A injections: adverse events reported to the US Food and Drug Administration in therapeutic and cosmetic cases. J Am Acad Dermatol 53, 407-415 (2005).

8. Dong, M., Tepp, W.H., Liu, H., Johnson, E.A. & Chapman, E.R. Mechanism of botulinum neurotoxin B and G entry into hippocampal neurons. J Cell Biol 179, 1511-1522 (2007).

9. Peng, L., Tepp, W.H., Johnson, E.A. & Dong, M. Botulinum neurotoxin D uses synaptic vesicle protein SV2 and gangliosides as receptors. PLoS Pathog 7, el 002008 (2011).

10. Dong, M., et al. Synaptotagmins I and II mediate entry of botulinum neurotoxin B into cells. J Cell Biol 162, 1293-1303 (2003).

11. Nishiki, T., et al. Identification of protein receptor for Clostridium botulinum type B neurotoxin in rat brain synaptosomes. J Biol Chem 269, 10498-10503 (1994).

12. Rummel, A., Karnath, T., Henke, T., Bigalke, H. & Binz, T. Synaptotagmins I and II act as nerve cell receptors for botulinum neurotoxin G. J Biol Chem 279, 30865-30870 (2004). 13. Peng, L., et al. Botulinum neurotoxin D-C uses synaptotagmin I/II as receptors and human synaptotagmin II is not an effective receptor for type B, D-C, and G toxins. J Cell Sci (2012).

14. Dong, M., et al. SV2 is the protein receptor for botulinum neurotoxin A. Science 312, 592-596 (2006).

15. Dong, M., et al. Glycosylated SV2A and SV2B mediate the entry of botulinum neurotoxin E into neurons. MolBiol Cell 19, 5226-5237 (2008).

16. Mahrhold, S., Rummel, A., Bigalke, H., Davletov, B. & Binz, T. The synaptic vesicle protein 2C mediates the uptake of botulinum neurotoxin A into phrenic nerves. FEBS Lett 580, 2011-2014 (2006).

17. Rummel, A., et al. Botulinum neurotoxins C, E and F bind gangliosides via a conserved binding site prior to stimulation-dependent uptake with botulinum neurotoxin F utilising the three isoforms of SV2 as second receptor. J Neurochem 110, 1942-1954 (2009).

18. Fu, Z., Chen, C, Barbieri, J.T., Kim, J.J. & Baldwin, M R. Glycosylated SV2 and gangliosides as dual receptors for botulinum neurotoxin serotype F. Biochemistry 48, 5631- 5641 (2009).

19. Montecucco, C. How do tetanus and botulinum toxins bind to neuronal membranes? TIBS, 314-317 (1986).

20. Nishiki, T., et al. The high-affinity binding of Clostridium botulinum type B neurotoxin to synaptotagmin II associated with gangliosides GT lb/GDI a. FEBS Lett 378, 253-257 (1996).

21. Pang, Z.P., et al. Synaptotagmin-2 is essential for survival and contributes to Ca2+ triggering of neurotransmitter release in central and neuromuscular synapses. JNeurosci 26, 13493-13504 (2006).

22. Strotmeier, J., Willjes, G., Binz, T. & Rummel, A. Human synaptotagmin-II is not a high affinity receptor for botulinum neurotoxin B and G: increased therapeutic dosage and immunogenicity. FEBS Lett 586, 310-313 (2012).

23. Craxton, M. A manual collection of Syt, Esyt, Rph3a, Rph3al, Doc2, and Dblc2 genes from 46 metazoan genomes—an open access resource for neuroscience and evolutionary biology. BMC Genomics 11, 37 (2010).

24. Brin, M.F., et al. Safety and efficacy of NeuroBloc (botulinum toxin type B) in type A-resistant cervical dystonia. Neurology 53, 1431-1438 (1999). 25. Pappert, E.J. & Germanson, T. Botulinum toxin type B vs. type A in toxin-naive patients with cervical dystonia: Randomized, double-blind, noninferiority trial. Mov Disord 23, 510-517 (2008).

26. Wang, J., et al. Longer-acting and highly potent chimaeric inhibitors of excessive exocytosis created with domains from botulinum neurotoxin A and B. Biochem J 444, 59-67 (2012).

27. Rummel, A., Mahrhold, S., Bigalke, H. & Binz, T. Exchange of the H(CC) domain mediating double receptor recognition improves the pharmacodynamic properties of botulinum neurotoxin. FEBSJ27S, 4506-4515 (2011).

28. Kozaki, S., et al. Characterization of Clostridium botulinum type B neurotoxin associated with infant botulism in japan. Infect Immun 66, 4811-4816 (1998).

29. Ihara, H., et al. Sequence of the gene for Clostridium botulinum type B neurotoxin associated with infant botulism, expression of the C-terminal half of heavy chain and its binding activity. Biochim Biophys Acta 1625, 19-26 (2003).

30. Rummel, A., Mahrhold, S., Bigalke, H. & Binz, T. The HCC-domain of botulinum neurotoxins A and B exhibits a singular ganglioside binding site displaying serotype specific carbohydrate interaction. Mol Microbiol 51, 631-643 (2004).

31. Chai, Q., et al. Structural basis of cell surface receptor recognition by botulinum neurotoxin B. Nature 444, 1096-1100 (2006).

32. Jin, R., Rummel, A., Binz, T. & Brunger, A.T. Botulinum neurotoxin B recognizes its protein receptor with high affinity and specificity. Nature 444, 1092-1095 (2006).

33. Arnon, S.S., et al. Botulinum toxin as a biological weapon: medical and public health management. Jama 2S5, 1059-1070 (2001).

34. Moriishi, K., et al. Mosaic structures of neurotoxins produced from Clostridium botulinum types C and D organisms. Biochim Biophys Acta 1307, 123-126 (1996).

35. Hill, K.K., et al. Genetic diversity among Botulinum Neurotoxin-producing clostridial strains. J Bacteriol 189, 818-832 (2007).

36. Lalli, G., et al. Functional characterisation of tetanus and botulinum neurotoxins binding domains. J Cell Sci 112 ( Pt 16), 2715-2724 (1999).

[00148] The present invention may be as defined in any one of the following numbered paragraphs. A botulinum neurotoxin (BoNT) polypeptide comprising:

a) a protease domain;

b) a protease cleavage site;

c) a translocation domain; and

d) a modified receptor binding domain of Clostridial botulinum serotype B (B-H c ), comprising one or more substitution mutations corresponding to substitution mutations in serotype B, strain 1, selected from the group consisting of:

E1191C, E1191V, E1191L, E1191Y, S1199W, S1199E, SI 199H, Wl 178Y, Wl 178Q, Wl 178A, Wl 178S, Yl 183C, Yl 183P and combinations thereof. The BoNT polypeptide of paragraph 1, wherein the modified (B-H c ) comprises one substitution mutation corresponding to a substitution mutation in serotype B, strain 1, selected from the group consisting of E1191C, El 191V, E1191L, El 191 Y, S1199E, SI 199H, Wl 178Y, Wl 178Q, Wl 178A, Wl 178S, Yl 183C, and Yl 183P. The BoNT polypeptide of paragraph 2, wherein the modified (B-H c ) comprises the substitution mutation corresponding to El 191C in serotype B, strain 1. The BoNT polypeptide of paragraph 2, wherein the modified (B-H c ) comprises the substitution mutation corresponding to El 191V in serotype B, strain 1. The BoNT polypeptide of paragraph 1, wherein the modified (B-H c ) comprises two substitution mutations. tulinum neurotoxin (BoNT) polypeptide comprising:

a) a protease domain;

b) a protease cleavage site;

c) a translocation domain; and

d) a modified receptor binding domain of Clostridial botulinum serotype B (B-H c ), comprising two or more substitution mutations corresponding to substitution mutations in serotype B, strain 1, wherein one of the substitution mutations is selected from the group consisting of: E1191Q, E1191M, E1191C, E1191V, E1191L, and E1191Y. The BoNT polypeptide of paragraph 6, wherein one other of the substitution mutations corresponds to S1199W, S1199E, S1199H, S1199Y, W1178Y, W1178Q, W1178A, W1178S, Y1183C, Y1183P, S1199F or S1199L in serotype B, strain 1. The BoNT polypeptide of any one of paragraphs 5 - 7, wherein the two substitution mutations correspond to El 191M and SI 199W, El 191M and Wl 178Q, El 191C and S1199W; E1191C and S1199Y, El 191C and Wl 178Q, El 191Q and SI 199W,

El 191 V and SI 199W, El 191 V and SI 199Y, or El 191 V and Wl 178Q, in serotype B, strain 1. The BoNT polypeptide of any one of paragraphs 5 - 8, wherein the two substitution mutations correspond to El 191M and SI 199W in serotype B, strain 1. The BoNT polypeptide of one of paragraphs 5 - 8, wherein the two substitution mutations correspond to El 191M and Wl 178Q in serotype B, strain 1. The BoNT polypeptide of one of paragraphs 5 - 8, wherein the two substitution mutations correspond to El 191C and SI 199W in serotype B, strain 1. The BoNT polypeptide of one of paragraphs 5 - 8, wherein the two substitution mutations correspond to El 191C and SI 199 Y in serotype B, strain 1. The BoNT polypeptide of one of paragraphs 5 - 8, wherein the two substitution mutations correspond to El 191C and Wl 178Q in serotype B, strain 1. The BoNT polypeptide of one of paragraphs 5 - 8, wherein the two substitution mutations correspond to El 191Q and SI 199W in serotype B, strain 1. The BoNT polypeptide of one of paragraphs 5 - 8, wherein the two substitution mutations correspond to El 191V and SI 199W in serotype B, strain 1. The BoNT polypeptide of one of paragraphs 5 - 8, wherein the two substitution mutations correspond to El 191V and SI 199 Y in serotype B, strain 1. The BoNT polypeptide of one of paragraphs 5 - 8, wherein the two substitution mutations correspond to El 191V and Wl 178Q in serotype B, strain 1. The BoNT polypeptide of paragraph 6, wherein the modified (B-H c ) comprises three substitution mutations. The BoNT polypeptide of paragraph 18, wherein the three substitution mutations are at positions that correspond to El 191, Y1183 and SI 199 or to El 191, SI 199 and W1178 of serotype B, strain 1. The BoNT polypeptide of paragraph 19, wherein the three substitution mutations correspond to E1191M, S1199W and W1178Q of serotype B, strain 1. A botulinum neurotoxin (BoNT) polypeptide comprising:

a) a protease domain;

b) a protease cleavage site;

c) a translocation domain; and

d) a modified receptor binding domain of Clostridial botulinum serotype B (B-H c ), comprising a substitution mutation at a position corresponding to SI 199 or SI 201 of serotype B, strain 1. The BoNT polypeptide of any one of paragraphs 1 -21, wherein the modified B-H c is of strain 1. The BoNT polypeptide of any one of paragraphs 1-22 wherein the protease domain, translocation domain, and protease cleavage site are from serotype selected from the group consisting of A, B, C, D, E, F, G, and combinations thereof. The BoNT polypeptide of paragraph 23, wherein the protease domain, translocation domain, and protease cleavage site are from serotype B, strain 1. The BoNT polypeptide of paragraph 23, wherein the protease domain, translocation domain, and protease cleavage site are from serotype A, strain 1. A polypeptide comprising a modified receptor binding domain of Clostridial botulinum serotype B (B-H c ) comprising one or more substitution mutations corresponding to substitution mutations in serotype B, strain 1, selected from the group consisting of:

E1191C, E1191V, E1191L, E1191Y, S1199W, S1199E, SI 199H, Wl 178Y, Wl 178Q, Wl 178A, Wl 178S, Yl 183C, Yl 183P and combinations thereof.

The polypeptide of paragraph 26, wherein the modified (B-H c ) compri

substitution mutations.

A polypeptide comprising a modified receptor binding domain of Clostridial botulinum serotype B (B-H c ) comprising two or more substitution mutations corresponding to substitution mutations in serotype B, strain 1, wherein one of the substitution mutations is selected from the group consisting of El 191Q, El 191M, E1191C, E1191V, E1191L, and E1191Y.

The polypeptide of paragraph 26, wherein one of the substitution mutations corresponds to S1199W, S1199E, S1199H, SI 199Y, Wl 178Y, Wl 178Q, Wl 178 A, W1178S, Y1183C, Y1183P, S1199F or S1199L in serotype B, strain 1.

The polypeptide of any one of paragraphs 27 - 29, wherein the two substitution mutations correspond to El 191M and SI 199W, El 191M and Wl 178Q, El 191C and S1199W; E1191C and S1199Y, El 191C and Wl 178Q, El 191Q and SI 199W, El 191 V and SI 199W, El 191 V and SI 199Y, or El 191 V and Wl 178Q, in serotype B, strain 1. The polypeptide of any one of paragraphs 27-30, wherein the two substitution mutations correspond to El 191M and SI 199W in serotype B, strain 1. The polypeptide of one of paragraphs 27-30, wherein the two substitution mutations correspond to El 191M and Wl 178Q in serotype B, strain 1. The polypeptide of one of paragraphs 27-30, wherein the two substitution mutations correspond to El 191C and SI 199W in serotype B, strain 1. The polypeptide of one of paragraphs 27-30, wherein the two substitution mutations correspond to El 191C and SI 199Y in serotype B, strain 1. The polypeptide of one of paragraphs 27-30, wherein the two substitution mutations correspond to El 191C and Wl 178Q in serotype B, strain 1. The polypeptide of one of paragraphs 27-30, wherein the two substitution mutations correspond to El 191Q and SI 199W in serotype B, strain 1. The polypeptide of one of paragraphs 27-30, wherein the two substitution mutations correspond to El 191V and SI 199W in serotype B, strain 1. The polypeptide of one of paragraphs 27-30, wherein the two substitution mutations correspond to El 191V and SI 199 Y in serotype B, strain 1. The polypeptide of one of paragraphs 27-30, wherein the two substitution mutations correspond to El 191V and Wl 178Q in serotype B, strain 1. The polypeptide of paragraph 30, wherein the modified (B-H c ) comprises three substitution mutations. The polypeptide of paragraph 40, wherein the three substitution mutations are at positions that correspond to E 1191 , Yl 183 and S 1199 or to E 1191 , S 1199 and W 1178 of serotype B, strain 1. The polypeptide of paragraph 41, wherein the three substitution mutations correspond to E1191M, S1199W and W1178Q of serotype B, strain 1. The polypeptide of any one of paragraphs 26 - 42, wherein the modified B-H c is of strain 1. A chimeric molecule comprising a first portion that is a modified receptor binding domain of Clostridial botulinum serotype B (B-H c ) linked to a second portion, wherein the modified B-H c comprises one or more substitution mutations selected from the group consisting of:

E1191C, E1191V, E1191L, E1191Y, S1199W, S1199E, SI 199H, Wl 178Y, Wl 178Q, Wl 178A, Wl 178S, Yl 183C, Yl 183P and combinations thereof. The chimeric molecule of paragraph 44, wherein the modified B-H c comprises two substitution mutations. A chimeric molecule comprising a first portion that is a modified receptor binding domain of Clostridial botulinum serotype B (B-H c ) linked to a second portion, wherein the modified B-H c comprises two or more substitution mutations

corresponding to substitution mutations in serotype B, strain 1, wherein one of the substitution mutations is selected from the group consisting of:

E1191Q, E1191M, E1191C, E1191V, E1191L, and E1191Y. The chimeric molecule of paragraph 46, wherein one of the substitution mutations corresponds to S1199W, S1199E, S1199H, SI 199Y, Wl 178Y, Wl 178Q, Wl 178 A, W1178S, Y1183C, Y1183P, S1199F or S1199L in serotype B, strain 1. The chimeric molecule of any one of paragraphs 45 - 47, wherein the two substitution mutations correspond to El 191M and SI 199W, El 191M and Wl 178Q, El 191C and S1199W; E1191C and S1199Y, El 191C and Wl 178Q, El 191Q and SI 199W,

El 191 V and SI 199W, El 191 V and SI 199Y, or El 191 V and Wl 178Q, in serotype B, strain 1. The chimeric molecule of any one of paragraphs 45 - 47, wherein the two substitution mutations correspond to El 191M and SI 199W in serotype B, strain 1. The chimeric molecule of any one of paragraphs 45 - 47, wherein the two substitution mutations correspond to El 191M and Wl 178Q in serotype B, strain 1. The chimeric molecule of any one of paragraphs 45 - 47, wherein the two substitution mutations correspond to El 191C and SI 199W in serotype B, strain 1. The chimeric molecule of any one of paragraphs 45 - 47, wherein the two substitution mutations correspond to El 191C and SI 199 Y in serotype B, strain 1. The chimeric molecule of any one of paragraphs 45 - 47, wherein the two substitution mutations correspond to El 191C and Wl 178Q in serotype B, strain 1. The chimeric molecule of any one of paragraphs 45 - 47, wherein the two substitution mutations correspond to El 191Q and SI 199W in serotype B, strain 1. The chimeric molecule of any one of paragraphs 45 - 47, wherein the two substitution mutations correspond to El 191V and SI 199W in serotype B, strain 1. The chimeric molecule of any one of paragraphs 45 - 47, wherein the two substitution mutations correspond to El 191V and SI 199 Y in serotype B, strain 1. The chimeric molecule of any one of paragraphs 45 - 47, wherein the two substitution mutations correspond to El 191V and Wl 178Q in serotype B, strain 1. The chimeric molecule of one of paragraph 45 or 46, wherein the modified (B-H c ) comprises three substitution mutations. The chimeric molecule of paragraph 58, wherein the three substitution mutations are at positions that correspond to El 191, Y1183 and SI 199 or to El 191, SI 199 and W1178 of serotype B, strain 1. The chimeric molecule of paragraph 59, wherein the three substitution mutations correspond to E1191M, S1199W and W1178Q of serotype B, strain 1. The chimeric molecule of any one of paragraphs 44- 60, wherein the modified B-H c is of strain 1. The chimeric molecule of any one of paragraphs 44-61, wherein the first portion and the second portion are linked covalently. The chimeric molecule of any one of paragraphs 46-61, wherein the first portion and the second portion are linked non-covalently. The chimeric molecule of any one of paragraphs 44-61 wherein the second portion is selected from the group consisting of a small molecule, a nucleic acid, a short polypeptide and a protein. The chimeric molecule of paragraph 64, wherein the second portion is a bioactive molecule. The chimeric molecule of paragraph 64 or 65, wherein the second portion is a therapeutic polypeptide or non-polypeptide drug. The BoNT polypeptide, polypeptide or chimeric molecule of any one of paragraphs 1- 66 that exhibits significantly enhanced binding of the modified B-H c to human Sytll and/or significantly reduced binding of the modified B-H c to human Syt I as compared to an identical molecule lacking the substitution mutation(s). The BoNT polypeptide, polypeptide or chimeric molecule of any one of paragraphs 1- 66 wherein the substitution mutation produces significantly enhanced binding to human Sytll and/or significantly enhanced binding to human Syt I as compared to an identical molecule lacking the substitution mutation(s). A nucleic acid comprising a nucleotide sequence that encodes the polypeptide or chimeric molecule of any one of paragraphs 1- 68.

A nucleic acid vector comprising the nucleic acid of paragraph 69. 71. A cell comprising the nucleic acid vector of paragraph 70 or the nucleic acid of paragraph 69.

72. A cell expressing the polypeptide or chimeric molecule of any one of paragraphs 1- 68

73. A pharmaceutical composition comprising the botulinum neurotoxin (BoNT)

polypeptide of any one of paragraphs 1-25, 67 or 68, or the chimeric molecule of any one of paragraphs 44-66, or the nucleic acid vector of paragraph 70 or the nucleic acid of paragraph 69.

74. The pharmaceutical composition of paragraph 73, further comprising a

pharmaceutically acceptable excipient.

75. A kit comprising a pharmaceutical composition of paragraph 73 or 74 and directions for therapeutic administration of the pharmaceutical composition.

76. A method to produce a botulinum neurotoxin (BoNT) polypeptide, the method

comprising the steps of culturing the cell of paragraph 72 under conditions wherein said BoNT polypeptide is produced.

77. The method of paragraph 76 further comprising one or more of the following steps:

- recovering the BoNT polypeptide from the culture,

- purifying the BoNT polypeptide,

- activating the BoNT polypeptide, and/or

- formulating the BoNT polypeptide.

78. A method for treating a condition associated with unwanted neuronal activity

comprising administering a therapeutically effective amount of the BoNT polypeptide of any one of paragraphs 1-25, 67 or 68 to a subject to thereby contact one or more neurons exhibiting unwanted neuronal activity, to thereby treat the condition.

79. The method of paragraph 78, wherein the condition is selected from the group

consisting of , spasmodic dysphonia, spasmodic torticollis, laryngeal dystonia, oromandibular dysphonia, lingual dystonia, cervical dystonia, focal hand dystonia, blepharospasm, strabismus, hemifacial spasm, eyelid disorder, cerebral palsy, focal spasticity and other voice disorders, spasmodic colitis, neurogenic bladder, anismus, limb spasticity, tics, tremors, bruxism, anal fissure, achalasia, dysphagia and other muscle tone disorders and other disorders characterized by involuntary movements of muscle groups, lacrimation, hyperhydrosis, excessive salivation, excessive gastrointestinal secretions, secretory disorders, pain from muscle spasms, headache pain, migraine, and dermatological or aesthetic/cosmetic conditions.

80. The botulinum neurotoxin (BoNT) polypeptide of any one of paragraphs 1-25, 67 or 68, the pharmaceutical composition of one of paragraphs 73 or 74 or the chimeric molecule of any one of paragraphs 44-66, or the polypeptide of any one of paragraphs 26-43, for use in medicine.

81. The botulinum neurotoxin (BoNT) polypeptide of any one of paragraphs 1-25, 67 or 68, the pharmaceutical composition of one of paragraphs 73 or 74, or the chimeric molecule of any one of paragraphs 44-66, or the polypeptide of any one of paragraphs 67-43, for use in treating a condition associated with unwanted neuronal activity.

82. A method for identifying a modified receptor binding domain of botulinum

neurotoxin for binding to a receptor comprising;

a) expressing the modified receptor binding domain as a first fusion protein with T18 subunit of bacterial adenylate cyclase in a 2-hybrid assay, and expressing the receptor as a second fusion protein with T25 subunit of bacterial adenylate cyclase in the 2-hybrid assay;

b) analyzing a clonal E. coli colony that expresses both the first and second fusion protein for the presence of a positive indication above a negative control; and

c) identifying the modified receptor binding domain expressed by the colony exhibiting the positive indication as binding to the receptor.

83. The method of paragraph 82 wherein the positive indication is color development. 84. The method of any one of paragraphs 82 - 83, further comprising the step of analyzing the colony for the positive indication against a weakly positive control, and further identifying the modified receptor binding domain expressed by the colony exhibiting above the weakly positive control as binding to the receptor with high affinity.

85. The method of paragraph 82-84 that is performed with a library of modified receptor binding domains each expressed within respective colonies.

86. The method of any one of paragraphs 82-85 wherein the receptor is human.

87. The method of any one of paragraphs 82-86 wherein the modified receptor binding domain comprises one or more substitution mutations, wherein one of the substitution mutations corresponds to a mutation in serotype B, strain 1, selected from the group consisting of: El 191M, El 191 Q, El 191C, El 191 V, El 191L, El 191 Y, SI 199W, S1199E, S1199H, S1199F, SI 199L Wl 178Y, Wl 178Q, Wl 178A, Wl 178S, Yl 183C, Y1183P and S1199Y.

[00149] The invention is further illustrated by the following examples, which should not be construed as further limiting.

EXAMPLES

Identification of single mutations that enhance BoNT/B binding to h-Syt II

[00150] The co-crystal structure of BoNT/B bound to rat Syt II luminal domain has been solved by two studies in 2006 25 ' 26 . The structural information provided a sound basis for us to focus on limited numbers of residues within the binding interface for rational design mutagenesis studies. The conserved phenylalanine at position 54 forms multiple hydrophobic contacts with BoNT/B. Because leucine (in humans) is also hydrophobic, disruption of BoNT/B binding is likely due to size/shape differences between phenylalanine and leucine. The key was to identify possible changes in BoNT/B He region that may accommodate the change from phenylalanine to leucine.

[00151] All residues in BoNT/B that contribute to interactions between BoNT/B and Syt II were examined. These residues were well-defined by the BoNT/B-Syt II co-crystal structure, including Kl l 13, Dl 115, SI 116, PI 117, VI 118, W1178, Y1181, Yl 183, El 191, Kl 192, F1194, A1196, PI 197, SI 199, S1201, E1203, F1204, E1245, and Y1256 (Fig. 3), a total of 19 positions. The strategy was to first systematically replace the residues at each of these 19 positions with all other 19 possible amino acids and then test binding of these mutated BoNT/B-Hc to h-Syt II. Therefore, a total of 19 xl9 = 361 single point mutations needed to be generated and tested.

[00152] A bacterial adenylate cyclase two hybrid system (BACTH) was used to generate these 361 mutations and test their binding to h-Syt II as described in Fig. 4. Briefly, wild type (WT) BoNT/B-Hc was subcloned into a vector in frame with the split fragment (T18) of the bacterial adenylate cyclase. This T18-BoNT/B-H c fusion construct was amplified by PCR with primers harboring random tri-nucleotides (NNN) at the selected position in BoNT/B-H c (Fig. 4A). This generated a pool of constructs that encode all 20 different amino acids at the select site. This pool of constructs were then co-transformed into bacteria (E.coli strain BTHlOl), together with a construct that expresses h-Syt II (1-87) fused with the other half of the split bacterial adenylate cyclase (T25). Binding of a mutant BoNT/B-H c to h-Syt II brought T18 and T25 together and recovered the activity of adenylate cyclase, which lead to expression of lacZ gene and results in blue colonies on X-gel plates (Fig. 4B). The specific mutations introduced into BoNT/B-H c were identified by extracting and sequencing the constructs from these blue colonies.

[00153] Using this BACTH method, all 19 selected sites were screened in BoNT/B-H c . The numbers of total colonies and blue colonies for each site are listed in Figure 5A. Greater than 380 total colonies were counted for each position. The total colony number determined the possibility of covering all 20 amino acids at the selected mutation site. This was calculated by Clark-Carbon equation: P=l-(l-f) N , where f reflects the number of possible residues (f = 1/20 here as there are 20 different amino acids), and N is the total number of colonies. With a minimal number of 380 colonies, the probability of covering all 20 amino acids at a position is 99.8%. Therefore, it is likely that all 20 possible residues were covered once the number of colonies on the plates was over 380..

[00154] As shown in Figure 5 A, four positions were found to result in blue colonies, including El 191 (22.7%), W11178 (7.8%), Y1183 (4.0%), and SI 199 (5.1%). Among these four positions, El 191 had the highest levels of blue colonies. In addition, the blue colonies from El 191 also showed deeper blue color as compared to the other three sites, which suggested that the interaction between BoNT/B-Hc and h-Syt II could be stronger with El 191 mutations. Plasmids were extracted and sequenced from these blue colonies. The identified residues at each site are listed in Figure 5B: El 191M/C/V/Q/L/Y, Yl 183/C/P, S 1199W/E/Y/H, W 1178 Y/Q/A/S .

[00155] The interactions between human Syt II and those identified mutants were further verified by measuring the levels of β-galactosidase produced in bacteria, which was proportional to the total level of adenylate cyclase activity reconstituted by interactions between T18-BoNT/B-H c and T25-h-Syt II. Four mutations at El 191 sites, El 191M/C/V/Q, showed the strongest β-galactosidase activity among all mutations tested (Figure 6A), suggesting that replacing El 191 with one of these four residues significantly enhanced binding of T18-BoNT/B-H c to T25-h-Syt II.

[00156] Binding of mutant BoNT/B-H c to h-Syt II was further analyzed using pull-down assay as an alternative approach (Figure 6B). Briefly, GST-tagged mouse Syt II luminal domain (m-Syt II) and human Syt II luminal domain (h-Syt II) were immobilized on GST beads and were used to pull down mutant BoNT/B-H c expressed in bacteria. Binding of BoNT/B-Hc to GST-tagged Syt II was detected via immunoblot analysis, detecting the HA tag fused to BoNT/B-Hc. As shown in Figure 6B, El 191M/C/V/Q resulted in significant levels of binding to h-Syt II, which was consistent with the finding that these four mutations also displayed the strongest β-galactosidase activity (Figure 6A). Together, these results indicated that El 191M/C/V/Q are four primary mutations that gain the ability to bind h-Syt II robustly.

Combinational mutations in HCB further enhanced its binding to h-Syt II

[00157] Whether binding of BoNT/B-H c El 191M/C/V/Q to h-Syt II can be further enhanced by including a secondary mutation at a different site was then explored. The 1183, 1199, and 1178 sites were focused on as the candidates for the secondary mutation sites, as these are the only three sites that also resulted in blue colonies (Figure 5). Using El 191M as the primary mutations, double mutations were generated combining El 191M with all other 10 residue changes identified in BACTH screening at 1183, 1199, and 1178 sites as indicated in Figure 5B. These 10 double mutations were analyzed for their ability to bind h-Syt II in pull-down assays as indicated in Figure 7A. Three of them, El 191M/S1199W, El 191M/S1199Y, and El 191M/W1178Q, resulted in significant binding to h-Syt II (Figure 7A). These results suggested that including SI 199W/Y and Wl 178Q as the secondary mutation site would further enhance binding of BoNT/B-H c El 191M/C/V/Q to h-Syt II. Together, these data indicated that there are four choices of primary mutation at El 191 site (M/C/V/Q) and three choice of secondary mutation sites (SI 199W/Y and Wl 178Q) (Figure 7B), and a combination of a primary mutation with a secondary mutation can further enhance binding to h-Syt II.

[00158] Combining El 191M with Yl 183C/P actually reduced binding to h-Syt II (Figure 7A). The Syt II binding interface of BoNT/B is composed of two hydrophobic pouches as previously reported. El 191 and Yl 183 are located in the same pouch, while SI 199 and Wl 178 are located in another. Because El 191 is spatially close to Yl 183, a potential structural conflict may occur when mutating both of them, which may explain why double mutations at these two positions reduced binding to h-Syt II.

[00159] Determination and comparison of binding affinities between mutated BoNT/B-H c to h-Syt II quantitatively was pursued, aiming to select the double mutations with the best binding affinity. The binding affinity (K D ) was determined using a well-established bio-layer interferometry assay (Figure 8A). Briefly, GST tagged Syt proteins were immobilized onto a probe. The probe was first exposed to purified BoNT/B-H c at different concentrations (association phase, Figure 8A), followed by washing steps (dissociation phase, Figure 8A). Binding of BoNT/B-Hc to GST tagged Syt increases the total molecular weight/size on the probe, which results in a shift in light reflection at the probe that can be detected and analyzed. Binding parameters such as association constant (K on ), dissociation constant (K off ), and apparent binding affinity (K D ) can be calculated from the association and dissociation curve detected as indicated in Figure 8A.

[00160] Using this assay, all combinations of four primary mutations (El 191M/C/Q/V) were systematically characterized with three secondary mutations (SI 199W/Y, Wl 178Q). In addition, a triple mutation, El 191M/S1199W/W 1178Q, was also generated and analyzed. Binding of WT BoNT/B-Hc to mouse Syt II (m-Syt II) was measured as a positive control, which showed a binding K D at 0.13 μΜ (Figure 8B). As expected, binding of WT BoNT/B- H c to h-Syt II was too weak to be reliably determined, with an estimated K D over the detection limit (> 20 μΜ). A single primary mutation El 191M yielded a binding K D at 6.7 μΜ, a significant improvement over WT BoNT/B-Hc. Double mutations that combined a primary mutation site with a secondary mutation sites further improved the binding affinity to as high as 0.59 μΜ (El 191 V/Sl 199Y). As expected, the majority of double mutations improved the binding affinity to h-Syt II, with K D between 0.59 μΜ to 4.3 μΜ.

El 191Q/W 1178Q was the only one that did not bind to h-Syt II. The reason is not known, but it is possible that this double mutation may have induced unexpected conformational changes of the protein. The triple mutation El 191M/S1199W/W1178Q showed almost the same binding affinity as the double mutation El 191M/S1199W, suggesting that adding the third mutation site may not further improve the binding affinity. Together, these data confirmed that all double mutations between El 191M/C/Q/V and S1199W/Y, Wl 178Q, with the exception of El 1191Q/W1178Q, resulted in mutant BoNT/B-H c that can bind to h-Syt II robustly.

HCB mutants showed enhanced binding to h-Syt I

[00161] In addition to Syt II, Syt I also functions as a receptor for BoNT/B. In order to achieve the highest possible binding to human neurons, the modified BoNT/B mutants should not affect binding to human Syt I. Ideally, they may even increase binding to Syt I. Indeed, it was found that El 191M significantly enhanced binding of BoNT/B-H c to h-Syt I, as robust binding could be detected without the presence of the lipid co-receptor gangliosides (Figure 9A). The binding affinity between selected double mutations and h-Syt I was measured using bio-layer interferometry assay. As shown in Figure 9B, C, double mutations

El 191M/S1199Y (B-H c MY) and El 191 V/Sl 199Y (B-H c VY) displayed K D at 2.9 μΜ and 5.82 μΜ, respectively, whereas the WT BoNT/B-Hc binding to h-Syt I was too weak to be reliably determined, with estimated K D over the detection limit ( >20 μΜ).

ΗΓΒΜΥ binds to h-Syt II on neuronal surfaces

[00162] We next examined whether HCB MY mutant may bind to h-Syt II on physiologically relevant neuronal surfaces. To this end, we utilized cultured rat cortical neurons as a neuron model, which expresses Syt I but not Syt II (Dong et al, The Journal of cell biology

179(7): 1511-1522 (2007)). Thus, knocking down Syt I generated neurons with no endogenous receptors. Expressing full-length h-Syt II in these Syt I KD neurons created "humanized" neurons with only h-Syt II as the toxin receptor as we previously described (Peng et al., J Cell Sci. 125: 3233-42(2012)). As expected, WT H C B bound strongly to rat neurons and the binding was abolished after knocking down endogenous Syt I. Expression of full-length m-Syt II, but not h-Syt II nor a m-Syt II containing F54L mutation, restored binding of WT HcB (Figure 10A). In contrast, HCB MY showed robust binding to neurons that express m-Syt II, h-Syt II, or m-Syt II (F54L), demonstrating that H C B MY gain the enhanced ability to bind h-Syt II on neuronal surfaces (Figure 10B). BoNT/B mutant displayed enhanced efficacy to block neurotransmission in humanized neurons

[00163] To address key question whether enhanced binding to h-Syt II translates to improved efficacy at functional levels in neurons, we produced full-length WT BoNT/B and the mutant toxin containing El 191M/S 1 199Y point mutations (ΒοΝΤ/Β Μ γ) recombinantly in E.coli. Humanized neurons were exposed to a gradient of WT or ΒοΝΤ/Β Μ γ toxins. Cleavage of VAMP2 was examined by immunoblotting analysis. As shown in Figure 1 1 A, more VAMP2 was cleaved in neurons exposed to ΒοΝΤ/Β Μ γ compared to neurons exposed to WT BoNT/B at each toxin concentration tested, indicating that ΒοΝΤ/Β Μ γ targeted and entered neurons more efficiently than WT toxin.

[00164] We next monitored neurotransmitter release by recording miniature inhibitory postsynaptic currents (mlPSCs) using whole-cell patch-clamp recording. The frequency of mlPSCs reflects the activity of neurotransmitter release in a population of neurons. Entry of BoNT/B into presynaptic terminals blocks release of neurotransmitter, thus reducing frequency of mlPSCs (Figure 1 IB). Humanized neurons were exposed to a gradient of WT BoNT/B or ΒοΝΤ/Β Μ γ· As shown in Figure 1 1C, ΒοΝΤ/Β Μ γ showed a greatly enhanced potency, with a half maximum inhibitory concentration (IC 50 ) ~1 1-fold lower than WT toxin: BONT/BMY can achieve the same level of blockage on neurotransmitter release with 1 1 -fold lower toxin concentrations compared to WT toxin. These data demonstrated that enhanced binding to human receptors resulted in increased efficacy of toxin at functional levels in neurons.

[00165] The BACTH method was used to screen all possible single mutations at all 19 key residues in BoNT/B-Hc that form the binding pocket for Syt II. Four positions were identified that can be mutated to increase the binding affinity to h-Syt II, with El 191 site as the primary site, and S I 199, Wl 178, and Yl 183 as the secondary mutation sites. Double mutations combining the primary and secondary mutation sites, were created and tested, and showed that combining El 191M/C/V/Q with S 1 199Y/W or Wl 178Q yield double mutations with strong binding affinity to h-Syt II and h-Syt I.

Discussion

[00166] By combining rationale design based on available co-crystal structure of the

BoNT/B-Syt II complex with the BACTH method that saturates all possible single point mutations at each of the selected target residues, a series of point mutations in BoNT/B was identified that can bind to h-Syt II. These point mutations were further examined in combinations, revealing double mutants that gained high-affinity binding to h-Syt II.

Importantly, full-length BoNT/B containing the designed mutation showed ~11-fold higher potency than WT BoNT/B on humanized neurons, demonstrating that enhanced binding to toxin receptors translates to higher potency at functional levels in neurons.

Materials and Methods

[00167] Materials and constructs. The following antibodies were purchased from indicated vendors: Synapsin I (Clone 46.1, Synaptic Systems), VAMP2 (Clone 69.1, Synaptic

Systems), HA (16B12, Covance), and β-tubulin III (abl8207, Abeam). Bovine mixed brain gangliosides were purchased from Matreya LLC (Pleasant Gap, PA) and were reconstituted in Tris-buffered saline (TBS: 20 mM Tris, 150 mM NaCl) as previously described (Peng et al., PLoS pathogens 7(3):el002008(2011)). The cDNA encoding H C B (residue 857-1291, Genbank: ACA46990.1) was codon optimized for E. coli expression and synthesized by Genscript Inc. (New Brunswick, NJ). The following cDNAs were generously provided by indicated groups: rat Syt I (T.C. Sudhof, Palo Alto, CA), mouse Syt II (M. Fukuda, Ibaraki, Japan), human Syt I (R.B. Sutton, Lubbock, TX). DNA encoding H C B was subcloned into pET28a vector, with both a His6 tag and a HA tag (YPYDVPDYA (SEQ ID NO: 12)) fused to its N-terminus. Mutations in ¾B were generated via PCR using Site-directed Mutagenesis Kit (Agilent Technologies, CA). GST tagged Syt I/II fragments and Syt II F54L mutant were described previously (Dong M, et al. The Journal of cell biology 162(7): 1293-1303(2003); Peng et al., J Cell Sci. 125: 3233-42 (2012); Dong M, et al. Science 312(5773):592-596 (2006)).

[00168] BACTH (bacterial adenylate cyclase two-hybrid assay): The BACTH assay was performed according to the manufacturer's instruction (Euromedex). Two compatible plasmids, pUT18C and pKT25 were selected for the screen. H-Syt II luminal domain (residues 1-80) was cloned into pKT25 to generate pKT25-h-Syt II. HCB was cloned in pUT18C for producing T18-HCB. HCB mutant libraries were created with primers containing random nucleotide triplets (NNN) at designated positions. Each library was co- transformed with the pKT25-h-Syt II plasmid into E. coli indicator strain BTH101 by electroporation and screened on LB agar plates containing 100 μg/ml Ampicillin, 50 μg/ml Kanamycin, 0.5 mM IPTG, and 40 μg/ml X-Gal. The plates were incubated at 30 °C for 64 hours. Plasmids were extracted from blue colonies and sequenced. The total colony number determined the possibility of covering all 20 amino acids at the selected mutation site. This was calculated by Clark-Carbon equation: P=l-(l-f) N , where f reflects the number of possible residues (f = 1/20 here as there are 20 different amino acids), and N is the total number of colonies. With a minimal number of 380 colonies in the assays, the probability of covering all 20 amino acids at each position is > 99.8%.

[00169] β-Galactosidase assay: E. coli BTH101 cells with expressed proteins of interest were inoculated into liquid LB medium containing ampicillin, kanamycin, and IPTG (0.5 mM). The culture was grown overnight at 37°C to reach the stationary phase. The OD 60 o of the culture grown overnight was recorded before harvesting. One milliliter of the culture grown overnight was centrifuged, and cell pellets were washed twice with PBS and suspended in an equal volume of Z buffer (60 mM Na2HP04, 40 mM Na 2 HP0 4 , 10 mM KC1, 1 mM MgS0 4 , and 20 mM dithiothreitol [DTT]). One hundred microliters of resuspended bacterial cells was diluted in 1 ml of Z buffer (dilution factor [DF]=10). Afterwards, 100 ml of chloroform and 50 ml of 0.1% SDS were added and mixed well to permeabilize the cells. Two hundred fifty microliters of the mixture was then transferred into a new Microfuge tube and brought to 28°C, 50 ml of pre-warmed o-nitrophenyl-P-galactoside (4 mg/ml in Z buffer) was added, and the mixture was incubated at 28°C until a yellow color developed. The reaction was stopped by addition of 200 ml of 1 M Na 2 C0 3 . The A 420 and the precise time period of the reaction in minutes (T) were recorded, β-galactosidase activity was defined as (1000 χ A 420 χ DF)/(T OD 600 ) in Miller units.

[00170] Protein expression and purification: WT and mutants of ΒοΝΤ/Β-¾ were expressed as His 6 tagged recombinant proteins in E. coli. Syt I/II fragments and mutants were expressed as GST tagged recombinant proteins in E. coli. Both GST-fusion and His 6 -fusion proteins were purified as previously described 9 , with the induction temperature at 20°C overnight with 0.25 mM IPTG.

[00171] GST pull-down assays: Two types of pull-down assays were carried out. The first series were used to screen binding of mutant BoNT/B-H c to GST-tagged mouse Syt II (m-Syt II) and a mutant mouse Syt II (F54L) that mimicking human Syt II sequence (designated as h- Syt II). Briefly, 6 ml of E. coli expressing BoNT/B ¾ were spin down, re-suspended in 800 μΐ TBS, sonicated, and then incubated with 2% Triton X-100 for 1 hr at 4°C. Samples were then spun down at maximal speed for 15 min in a microcentrifuge at 4°C. The supernatants were collected and were used for pull-down assays by incubating with 10 μg of Syt proteins immobilized on glutathione-Sepharose beads (GE bioscience, Piscataway, NJ) at 4°C for 1 hr. Samples were washed three times in washing buffer (TBS with 0.5% Triton X-100), and analyzed by immunoblotting BoNT/B-Hc using the anti-HA antibody. For mutants with enhanced binding to h-Syt II, further pull-down assays were carried out by purifying these BoNT/B-Hc mutants as His6 tagged proteins as described previously 9 . Pull-down assays were then carried out using immobilized Syt fragments in 100 μΐ TBS buffer plus 0.5% Triton X- 100, with or without gangliosides (60 μg/ml), for 1 hr at 4 °C. Beads were washed three times using TBS buffer plus 0.5% Triton X-100. Ten percent of bound materials were subjected to SDS-PAGE followed immunoblot analysis.

[00172] Biolayer interferometry assay. The binding affinities between ¾Β variants and Syt I/Syt II were measured by BLI assay with the Blitz system (ForteBio). Briefly, the GST- tagged Syt I or Syt II (20 μg/ml) were immobilized onto Dip and Read™ Anti-GST

Biosensors (ForteBio) and balanced with PBS buffer. The biosensors were then exposed to series concentrations of ¾B, followed by washing with PBS. Binding affinities (K D ) were calculated using the Blitz system software following manufacture's instruction (ForteBio).

[00173] Neuron culture, lentivirus, and toxin binding/entry assay. Rat cortical neurons were prepared from E18-19 embryos as described previously (Peng et al. PLoS pathogens

7(3):el002008 (2011)). Constructs for Syt I KD, mSyt II, and h-Syt II expression in neurons was previously described (Peng et al., J Cell Sci. 125: 3233-42(2012)). Lentiviruses were added to neuron cultures at DIV5 (days in vitro), and toxin binding/entry experiments were carried out on DTV12-14. Toxins were diluted in high K + buffer (87 mM NaCl, 56 mM KCl, 1.5 mM KH 2 P0 4 , 8 mM Na 2 HP0 4 , 0.5 mM MgCl 2 , and 1 mM CaCl) and pre-warmed to 37 °C. Neurons were exposed to above toxin-containing buffers for 5 minutes at 37 °C followed by washing with PBS. These neurons were either subjected to immunostaining analysis, or incubated in toxin-free medium for additional 24 hours, followed by immunoblotting analysis.

[00174] mlPSC recording. Whole-cell patch-clamp recordings were made from DIV 14-18 cultured cortical neurons (DIV 14-18). The pipette solution contained (in mM): 135 CsCl, 10 HEPES, 1 EGTA, 1 Na-GTP, 4 Mg-ATP and 10 QX-314 (pH 7.4, adjusted with CsOH). The resistance of pipettes filled with intracellular solution varied between 4 and 5 ΜΩ. After formation of the whole-cell configuration and equilibration of the intracellular pipette solution, the series resistance was adjusted to 10 ΜΩ. Synaptic currents were monitored with an EPC-10/2 amplifier (HEKA) at -70 mV holding potential. The bath solution contained (in mM): 140 NaCl, 5 KC1, 2 CaCl, 1 MgC12, 10 HEPES, 10 glucose (pH 7.4, adjusted with NaOH). Spontaneous inhibitory postsynaptic currents (sIPSCs) and evoked inhibitory postsynaptic currents (elPSCs) were pharmacologically inhibited by adding AMPA and

MDA receptor blockers CNQX and APV to the extracellular bath solution. Spontaneous miniature inhibitory postsynaptic currents (mlPSCs) were monitored in the presence of tetrodotoxin (TTX) to block action potentials. Data were analyzed using Clampfit 10

(Molecular Devices), Origin8 software (Mocrocal Inc.), Mini Analysis software

(Synaptosoft), and Igor (Wavemetrics). Statistical analysis was performed with Student's t- test (*P < 0.01). All data shown are means ± S.E.M.s.

Conclusion

[00175] The above-presented results indicate new methods and compositions to improve binding of BoNT/B to its human receptors, and provide a way to create new generations of therapeutic BoNTs, by utilizing the modified receptor binding domain created in the present invention, with improved efficacy and specificity to target human neurons than the currently utilized WT BoNTs.

[00176] These efforts show that modifying the protein sequence of BoNT/B-Hc can create new versions that can bind human Syt II with high-affinity. Modification of the BoNT/B-H c protein sequence can be performed by either targeted mutagenesis (site-directed mutagenesis) or random mutagenesis of each amino acid residue within the region known for binding Syt I/II. These Syt binding regions are well defined by previous co-crystal structural studies 25 ' 26 . For instance, it composes of residues 1078-1291 in BoNT/Bl sequence (GenBank access No. : PI 0844). Although these studies were performed using BoNT/Bl sequence, all subtypes of BoNT/B can be used as the template to re-create the same or similar mutations as described here. Although the exact position for selected residues may not be identical in different BoNT/B subtypes, the analogous residues in the different BoNT/B subtypes can be easily identified (e.g., by sequence alignment when not at exactly the same position).

[00177] Specifically studied were all residues in the Syt II binding interface of BoNT/B based on reported BoNT/B-Syt II complex structure (PDB ID: 2NM1), including Kl 113, Dl 115, S1116, P1117, V1118, W1178, Y1181, Y1183, E1191, K1192, F1194, A1196, P1197, SI 199, S1201, E1203, F1204, E1245, and Y1256, as listed in Figure 3B. [00178] Mutagenesis is a common laboratory technique to create engineered protein products. Several methods to introduce mutations, including site-directed mutagenesis, random mutagenesis, combinatorial mutagenesis, and insertional mutagenesis, are available. Site- directed mutagenesis and random mutagenesis were applied to introduce mutations into BoNT/B-Hc sequence. Random mutagenesis is a powerful tool to create mutant libraries for screening. By using PCR primers with doped nucleotides, a pool of mutants was created containing substitutions to all other 19 amino acids at the positions listed in Figure 3B. The screening was then performed using BACTH system as illustrated in Figure 4.

[00179] Residues at position 1191, 1178, 1183, and 1199 were identified as important for substitution mutations to generate BoNT/B-H c mutants with enhanced binding to human Syt II. The specific substitutions were El 191M/C/V/Q/L/Y, Yl 183C/P, SI 199Y/W/E/H, W1178Y/Q/A/S (Figure 5A, B).

[00180] Further quantitative assays revealed that changing residue E1991 to M/C/V/Q resulted in the strongest binding to h-Syt II, as measured by β-galactosidase activity (Figure 6A) and pull-down assays (Figure 6B). Therefore, El 191 was identified as the primary site to introduce mutations that enhance binding to h-Syt II, and substitution mutations of

El 191/M/C/V/Q were identified as the primary mutations.

[00181] El 191M was used as the primary mutation to explore whether adding a secondary mutation can further enhance binding to h-Syt II (Figure 7A). Because Yl 183C/P,

S1199Y/W/E/H, and Wl 178Y/Q/A/S substitution mutations resulted in "light blue" colonies in BACTH screening (Figure 5B), these sites were selected as the potential secondary mutation site. Double mutations combining El 191M with these potential secondary mutations were generated and tested. Three mutations, SI 199W, SI 199 Y, and Wl 178Q were found to enhance binding to h-Syt II, when combined with the primary mutation at El 191M in pull-down assays (Figure 7A, B). These three substitution mutations were selected as secondary mutations.

[00182] Further quantitative assays were carried out to determine the binding affinity between BoNT/B-Hc mutants and h-Syt II, using bio-layer interferometry assay as described in Figure 8 A. Double mutations, with one from primary mutations (Ml 191M/C/Q/V) and the other from secondary mutations (SI 199W/Y, Wl 178Q), were measured for their binding affinities to h-Syt II (Figure 8B). The results showed that the following double mutations:

El 191M/C/Q/V combined with S1199W/Y, and El 191M/C/V combined with Wl 178Q have significantly enhanced binding affinities for h-Syt II (Figure 8B). These eleven double combinations were thus identified as mutations in BoNT/B-H c that enhance binding to h-Syt II the most.

[00183] Engineered BoNT/B-Hc mutants were found not only enhance binding to human Syt II, but also to human Syt I. El 191M/S1199Y and El 191 V/Sl 199Y were used to show that these mutants displayed significantly enhanced binding abilities to human Syt I as well as compared to WT BoNT/B-H c (Figure 9).

[00184] The modified BoNT/B-Hc mutants may contain amino acids substitutions at one or combinations of the amino acid residues E1191, Y1183, W1178 and SI 199 such as

E1191M/S1199W, El 191M/S1199Y, El 191M/W1178Q, El 191C/S1199W,

E1191C/S1199Y, El 191C/W1178Q, E1191Q/S1199W, El 191Q/S1199Y, El 191V/S1199W, E1191V/S1199Y, and El 191V/W1178Q.

[00185] Selected triple mutations by combining mutations at El 191/S1199/Wl 178 sites were also tested and found to have similar binding affinity as the double mutation at El 191/S1199 sites. For instance, E1191M/S1199W/W1178Q has a similar binding affinity as

El 191M/S1199W (Figure 8B). Therefore, triple mutations exhibited the enhanced binding affinity of the double mutations, although they did not seem to offer significant advantage over double mutations with respect to increased binding affinity.

[00186] These results indicate that polypeptides containing BoNT/B-H c with modified amino acid sequence related to the sequence of WT BoNT/B-Hc, wherein the modified BoNT/B-Hc has improved ability to bind human Syt I and II as compared to WT BoNT/B-Hc can be made and used therapeutically. The polypeptides may be, for example, in the form of full-length BoNT/B mutants, truncated BoNT/B mutants or recombined proteins that contain the same amino acid substitutions within the receptor binding domain (BoNT/B-Hc), as described above, and also, BoNT/B subtypes with amino acid substitutions at according positions. In addition, BoNT/B subtype can be modified to bind h-Syt II by replacing the residues within their receptor binding domain that are different from BoNT/Bl .

[00187] The full-length BoNT/B mutants encompassed contain amino acids substitutions at one or combinations (e.g., 2 or 3) of the amino acid residues El 191, Yl 183, Wl 178 and SI 199 such as E1191M/S1199W, El 191M/S1199Y, El 191M/W1178Q, El 191C/S1199W, E1191C/S1199Y, El 191C/W1178Q, E1191Q/S1199W, El 191Q/S1199Y, El 191V/S1199W, E1191V/S1199Y, and E1191V/W1178Q. The mutations can be made in the same manner as disclosed above for BoNT/B-Hc, using any one of BoNT/B subtypes as templates. These mutated BoNT/B toxins gain enhanced binding to both human Syt II and human Syt I, therefore will achieve higher efficacy and specificity to target human neurons than WT BoNT/B.

References for the Examples Section

1. Schiavo, G., Matteoli, M. & Montecucco, C. Neurotoxins affecting neuroexocytosis.

Physiol Rev 80, 717-766 (2000).

2. Johnson, E.A. Clostridial toxins as therapeutic agents: benefits of nature's most toxic proteins. Annu Rev Microbiol 53, 551-575 (1999).

3. Aoki, K.R. Botulinum toxin: a successful therapeutic protein. Curr Med Chem 11,

3085-3092 (2004).

4. Montecucco, C. & Molgo, J. Botulinal neurotoxins: revival of an old killer. Curr Opin Pharmacol 5, 274-279 (2005).

5. Lange, O., et al. Neutralizing antibodies and secondary therapy failure after treatment with botulinum toxin type A: much ado about nothing? Clin Neuropharmacol 32, 213-218 (2009).

6. Chapman, M.A., Barron, R., Tanis, D.C., Gill, C.E. & Charles, P.D. Comparison of botulinum neurotoxin preparations for the treatment of cervical dystonia. Clin Ther 29, 1325-1337 (2007).

7. Cote, T.R., Mohan, A.K., Polder, J.A., Walton, M.K. & Braun, M.M. Botulinum toxin type A injections: adverse events reported to the US Food and Drug Administration in therapeutic and cosmetic cases. J Am Acad Dermatol 53, 407-415 (2005).

8. Dong, M., Tepp, W.H., Liu, H., Johnson, E.A. & Chapman, E.R. Mechanism of

botulinum neurotoxin B and G entry into hippocampal neurons. J Cell Biol 179, 1511- 1522 (2007).

9. Peng, L., Tepp, W.H., Johnson, E.A. & Dong, M. Botulinum neurotoxin D uses

synaptic vesicle protein SV2 and gangliosides as receptors. PLoS Pathog 7, el 002008 (2011).

10. Dong, M., et al. Synaptotagmins I and II mediate entry of botulinum neurotoxin B into cells. J Cell Biol 162, 1293-1303 (2003).

11. Nishiki, T., et al. Identification of protein receptor for Clostridium botulinum type B neurotoxin in rat brain synaptosomes. J Biol Chem 269, 10498-10503 (1994).

12. Rummel, A., Karnath, T., Henke, T., Bigalke, H. & Binz, T. Synaptotagmins I and II act as nerve cell receptors for botulinum neurotoxin G. J Biol Chem 279, 30865- 30870 (2004). Peng, L., et al. Botulinum neurotoxin D-C uses synaptotagmin I/II as receptors and human synaptotagmin II is not an effective receptor for type B, D-C, and G toxins. J Cell Sci (2012).

Montecucco, C. How do tetanus and botulinum toxins bind to neuronal membranes? TIBS, 314-317 (1986).

Nishiki, T., et al. The high-affinity binding of Clostridium botulinum type B neurotoxin to synaptotagmin II associated with gangliosides GT lb/GDI a. FEBSLett 378, 253-257 (1996).

Pang, Z.P., et al. Synaptotagmin-2 is essential for survival and contributes to Ca2+ triggering of neurotransmitter release in central and neuromuscular synapses. J Neurosci 26, 13493-13504 (2006).

Strotmeier, J., Willjes, G., Binz, T. & Rummel, A. Human synaptotagmin-II is not a high affinity receptor for botulinum neurotoxin B and G: increased therapeutic dosage and immunogenicity. FEBSLett 586, 310-313 (2012).

Craxton, M. A manual collection of Syt, Esyt, Rph3a, Rph3al, Doc2, and Dblc2 genes from 46 metazoan genomes—an open access resource for neuroscience and evolutionary biology. BMC Genomics 11, 37 (2010).

Brin, M.F., et al. Safety and efficacy of NeuroBloc (botulinum toxin type B) in type A-resistant cervical dystonia. Neurology 53, 1431-1438 (1999).

Pappert, E. J. & Germanson, T. Botulinum toxin type B vs. type A in toxin-naive patients with cervical dystonia: Randomized, double-blind, noninferiority trial. Mov Disord 23, 510-517 (2008).

Wang, J., et al. Longer-acting and highly potent chimaeric inhibitors of excessive exocytosis created with domains from botulinum neurotoxin A and B. Biochem J 444, 59-67 (2012).

Rummel, A., Mahrhold, S., Bigalke, H. & Binz, T. Exchange of the H(CC) domain mediating double receptor recognition improves the pharmacodynamic properties of botulinum neurotoxin. FEBSJ 27S, 4506-4515 (201 1).

Dong, M., et al. SV2 is the protein receptor for botulinum neurotoxin A. Science 312, 592-596 (2006).

Arnon, S.S., et al. Botulinum toxin as a biological weapon: medical and public health management. Jama 2S5, 1059-1070 (2001).

Jin, R., Rummel, A., Binz, T. & Brunger, A.T. Botulinum neurotoxin B recognizes its protein receptor with high affinity and specificity. Nature 444, 1092-1095 (2006). Chai, Q., et al. Structural basis of cell surface receptor recognition by botulinum neurotoxin B. Nature 444, 1096-1100 (2006).




 
Previous Patent: SYNTHESIS OF DESOSAMINES

Next Patent: MULTIFUNCTION TORCH