Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
EXPRESSION VECTORS COMPRISING ENGINEERED GENES
Document Type and Number:
WIPO Patent Application WO/2018/229254
Kind Code:
A1
Abstract:
The invention provides expression vectors, nucleic acids, vector particles and methods of treatment involving these vector particles, comprising an engineered KCNA 1 gene encoding an edited Kv1.1 potassium channel, as well as methods of confirming the presence of engineered KCNA 1 mRNA in a cell. The features of the engineered KCNA 1 gene combine to advantageously enhance the translation and activity of the Kv1.1 protein and improve detection of KCNA 1 gene expression in a cell and can be used for example in the treatment of epilepsy and similar neurological disorders.

Inventors:
SCHORGE STEPHANIE (GB)
WALKER MATTHEW CHARLES (GB)
KULLMANN DIMITRI M (GB)
SNOWBALL ALBERT (GB)
CHABROL ELODIE (GB)
Application Number:
PCT/EP2018/065953
Publication Date:
December 20, 2018
Filing Date:
June 15, 2018
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
UCL BUSINESS PLC (GB)
International Classes:
C12N15/86; A61K38/17; C12Q1/6813
Domestic Patent References:
WO2015136247A12015-09-17
WO2008096268A22008-08-14
Other References:
TARUN BHALLA ET AL: "Control of human potassium channel inactivation by editing of a small mRNA hairpin", NAT. STRUCT. MOL. BIOL., vol. 11, no. 10, 1 October 2004 (2004-10-01), New York, pages 950 - 956, XP055492110, ISSN: 1545-9993, DOI: 10.1038/nsmb825
R. C. WYKES ET AL: "Optogenetic and Potassium Channel Gene Therapy in a Rodent Model of Focal Neocortical Epilepsy", SCIENCE TRANSLATIONAL MEDICINE, vol. 4, no. 161, 12 November 2012 (2012-11-12), pages 161ra152 - 161ra152, XP055185925, ISSN: 1946-6234, DOI: 10.1126/scitranslmed.3004190
ANNE KATHRIN STREIT ET AL: "RNA editing of Kv1.1 channels may account for reduced ictogenic potential of 4-aminopyridine in chronic epileptic rats : RNA Editing of Kv1.1 and 4-AP Resistance", EPILEPSIA, vol. 52, no. 3, 1 March 2011 (2011-03-01), NEW YORK, US, pages 645 - 648, XP055492144, ISSN: 0013-9580, DOI: 10.1111/j.1528-1167.2011.02986.x
DATABASE UniProt [online] 13 November 2013 (2013-11-13), "SubName: Full=Potassium voltage-gated channel subfamily A member 1 {ECO:0000313|EMBL:JAB52125.1};", XP002783058, retrieved from EBI accession no. UNIPROT:U3FYB1 Database accession no. U3FYB1
DATABASE UniProt [online] 7 March 2006 (2006-03-07), "SubName: Full=Potassium voltage-gated channel, shaker-related subfamily, member 1 {ECO:0000313|EMBL:AAI12971.1};", XP002783059, retrieved from EBI accession no. UNIPROT:Q2KHP0 Database accession no. Q2KHP0
WYKES R C ET AL: "LENTIVECTOR-MEDIATED POTASSIUM CHANNEL OVEREXPRESSION DECREASES FOCAL CORTICAL SEIZURE FREQUENCY", EPILEPSIA, vol. 58, no. Suppl. 5, Sp. Iss. SI, December 2017 (2017-12-01), & 32ND INTERNATIONAL EPILEPSY CONGRESS; BARCELONA, SPAIN; SEPTEMBER 02 -06, 2017, pages S25, XP002783060
SNOWBALL ALBERT ET AL: "Changing channels in pain and epilepsy: Exploiting ion channel gene therapy for disorders of neuronal hyperexcitability", FEBS LETTERS, ELSEVIER, AMSTERDAM, NL, vol. 589, no. 14, 12 May 2015 (2015-05-12), pages 1620 - 1634, XP029140791, ISSN: 0014-5793, DOI: 10.1016/J.FEBSLET.2015.05.004
SAMBROOK ET AL.: "Molecular Cloning: a Laboratory Manual, 2nd ed", 1989, COLD SPRING HARBOR LABORATORY PRESS
AUSUBEL ET AL.: "Current Protocols in Molecular Biology, 2nd ed", 1995, JOHN WILEY & SONS
HOOPENGARDNER ET AL., SCIENCE, vol. 301, no. 5634, 2003, pages 832 - 6
ZHANG ET AL., BRAIN RESEARCH, vol. 1377, 2011, pages 1 - 12
RASMUSSEN ET AL., BRAIN RESEARCH, vol. 1144, 2007, pages 19 - 32
LIU H. ET AL., JOURNAL OF NEUROSCIENCE, vol. 23, no. 18, 2003, pages 7143 - 54
PEEL AL. ET AL., GENE THERAPY, vol. 4, no. 1, 1997, pages 16 - 24
KESSLER MA. ET AL., BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH, vol. 112, no. i-2, 2003, pages 8 - 23
KESSLER MA. ET AL., BIOCHEMICAL & BIOPHYSICAL RESEARCH COMMUNICATIONS, vol. 288, no. 4, 2001, pages 809 - 18
YAWORSKY PJ. ET AL., JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 272, no. 40, 1997, pages 25112 - 20
KUGLER ET AL.: "Human synapsin 1 gene promoter confers highly neuron-specific long-term transgene expression from an adenoviral vector in the adult rat brain depending on the transduced area", GENE THERAPY, vol. 10, no. 4, 2003, pages 337 - 47, XP055332874, DOI: doi:10.1038/sj.gt.3301905
DISS ET AL., MOLECULAR AND CELLULAR NEUROSCIENCE, vol. 37, no. 3, 2008, pages 537 - 47
PUHL; IKEDA, JOURNAL OF NEUROCHEMISTRY, vol. 106, no. 3, 2008, pages 1209 - 24
SHAW; CORNETTA, BIOMEDICINES, vol. 2, no. 1, 2014, pages 14 - 35
DULL, T. ET AL.: "A Third-generation lentivirus vector with a conditional packaging system", J. VIROL, vol. 72, no. 11, 1998, pages 8463 - 71
KAFRI ET AL., J VIROL., vol. 73, no. 1, 1999, pages 576 - 584
YANEZ-MUNOZ ET AL., NATURE MEDICINE, vol. 12, no. 3, 2006, pages 348 - 53
RHO; SANKAR, EPILEPSIA, vol. 40, 1999, pages 1471 - 1483
BAUM, C.; KALLE, C.; STAAL, F.J.T.; LI, Z.; FEHSE, B.; SCHMIDT, M.; WEERKAMP, F.; KARLSSON, S.; WAGEMAKER, G.; WILLIAMS, D.A.: "Chance or necessity? Insertional mutagenesis in gene therapy and its consequences", MOL. THER. J. AM. SOC. GENE THER., vol. 9, 2004, pages 5 - 13
BHALLA, T.; ROSENTHAL, J.J.C.; HOLMGREN, M.; REENAN, R.: "Control of human potassium channel inactivation by editing of a small mRNA hairpin", NAT. STRUCT. MOL. BIOL., vol. 11, 2004, pages 950 - 956, XP055492110, DOI: doi:10.1038/nsmb825
BIFFI, A.; MONTINI, E.; LORIOLI, L.; CESANI, M.; FUMAGALLI, F.; PLATI, T.; BALDOLI, C.; MARTINO, S.; CALABRIA, A.; CANALE, S. ET A: "Lentiviral Hematopoietic Stem Cell Gene Therapy Benefits Metachromatic Leukodystrophy", SCIENCE, vol. 341, 2013, pages 1233158, XP008163598, DOI: doi:10.1126/science.1233158
BOVOLENTA, R.; ZUCCHINI, S.; PARADISO, B.; RODI, D.; MERIGO, F.; NAVARRO MORA, G.; OSCULATI, F.; BERTO, E.; MARCONI, P.; MARZOLA,: "Hippocampal FGF-2 and BDNF overexpression attenuates epileptogenesis-associated neuroinflammation and reduces spontaneous recurrent seizures", J. NEUROINFLAMMATION, vol. 7, 2010, pages 81, XP021079554, DOI: doi:10.1186/1742-2094-7-81
BURNS, J.C.; FRIEDMANN, T.; DRIEVER, W.; BURRASCANO, M.; YEE, J.K.: "Vesicular stomatitis virus G glycoprotein pseudotyped retroviral vectors: concentration to very high titer and efficient gene transfer into mammalian and nonmammalian cells", PROC. NATL. ACAD. SCI. U. S. A., vol. 90, 1993, pages 8033 - 8037, XP000674727, DOI: doi:10.1073/pnas.90.17.8033
CARTIER, N.; HACEIN-BEY-ABINA, S.; BARTHOLOMAE, C.C.; VERES, G.; SCHMIDT, M.; KUTSCHERA, I.; VIDAUD, M.; ABEL, U.; DAL-CORTIVO, L.: "Hematopoietic stem cell gene therapy with a lentiviral vector in X-linked adrenoleukodystrophy", SCIENCE, vol. 326, 2009, pages 818 - 823, XP055268270, DOI: doi:10.1126/science.1171242
D'ADAMO MC; CATACUZZENO L; DI GIOVANNI G; FRANCIOLINI F; PESSIA M: "K(+) channelepsy: progress in the neurobiology of potassium channels and epilepsy", FRONT. CELL. NEUROSCI., vol. 7, 2013, pages 134
DEVINSKY, O.: "Sudden, unexpected death in epilepsy", N. ENGL. J. MED., vol. 365, 2011, pages 1801 - 1811
DITTGEN, T.; NIMMERJAHN, A.; KOMAI, S.; LICZNERSKI, P.; WATERS, J.; MARGRIE, T.W.; HELMCHEN, F.; DENK, W.; BRECHT, M.; OSTEN, P.: "Lentivirus-based genetic manipulations of cortical neurons and their optical and electrophysiological monitoring in vivo", PROC. NATL. ACAD. SCI. U. S. A., vol. 101, 2004, pages 18206 - 18211, XP003003909, DOI: doi:10.1073/pnas.0407976101
GALANOPOULOU, A.S.; BUCKMASTER, P.S.; STALEY, K.J.; MOSHE, S.L.; PERUCCA, E.; ENGEL, J., JR; LOSCHER, W.; NOEBELS, J.L.; PITKANEN,: "Identification of new epilepsy treatments: issues in preclinical methodology", EPILEPSIA, vol. 53, 2012, pages 571 - 582
HABERMAN, R.P.; SAMULSKI, R.J.; MCCOWN, T.J.: "Attenuation of seizures and neuronal death by adeno-associated virus vector galanin expression and secretion", NAT MED, vol. 9, 2003, pages 1076 - 1080, XP002427952, DOI: doi:10.1038/nm901
HACEIN-BEY-ABINA, S.; VON KALLE, C.; SCHMIDT, M.; MCCORMACK, M.P.; WULFFRAAT, N.; LEBOULCH, P.; LIM, A.; OSBORNE, C.S.; PAWLIUK, R: "LM02-associated clonal T cell proliferation in two patients after gene therapy for SCID-X1", SCIENCE, vol. 302, 2003, pages 415 - 419, XP002987515, DOI: doi:10.1126/science.1088547
HEEROMA, J.H.; HENNEBERGER, C.; RAJAKULENDRAN, S.; HANNA, M.G.; SCHORGE, S.; KULLMANN, D.M.: "Episodic ataxia type 1 mutations differentially affect neuronal excitability and transmitter release", DIS. MODEL. MECH., vol. 2, 2009, pages 612 - 619, XP055492132, DOI: doi:10.1242/dmm.003582
HILL, E.; KALLONIATIS, M.; TAN, S.S.: "Glutamate, GABA and precursor amino acids in adult mouse neocortex: cellular diversity revealed by quantitative immunocytochemistry", CEREB. CORTEX N. Y. N 1991, vol. 10, 2000, pages 1132 - 1142
HOPPE, C.; ELGER, C.E.: "Depression in epilepsy: a critical review from a clinical perspective", NAT. REV. NEUROL., vol. 7, 2011, pages 462 - 472
KANTER-SCHLIFKE, I.; GEORGIEVSKA, B.; KIRIK, D.; KOKAIA, M.: "Seizure suppression by GDNF gene therapy in animal models of epilepsy", MOL. THER. J. AM. SOC. GENE THER., vol. 15, 2007, pages 1106 - 1113
THEODORE FRIEDMANN: "Advances in Genetics", 2014, ACADEMIC PRESS, article KANTOR, B.; MCCOWN, T.; LEONE, P.; GRAY, S.J.: "Chapter Two - Clinical Applications Involving CNS Gene Transfer", pages: 71 - 124
KATZEL, D.; NICHOLSON, E.; SCHORGE, S.; WALKER, M.C.; KULLMANN, D.M.: "Chemical-genetic attenuation of focal neocortical seizures", NAT. COMMUN., vol. 5, 2014, pages 3847
KULLMANN, D.M.; SCHORGE, S.; WALKER, M.C.; WYKES, R.C.: "Gene therapy in epilepsy-is it time for clinical trials?", NAT. REV. NEUROL., vol. 10, 2014, pages 300 - 304, XP055400192, DOI: doi:10.1038/nrneurol.2014.43
KWAN, P.; SCHACHTER, S.C.; BRODIE, M.J.: "Drug-resistant epilepsy", N. ENGL. J. MED., vol. 365, 2011, pages 919 - 926
LAVDAS, A.A.; MIONE, M.C.; PARNAVELAS, J.G: "Neuronal clones in the cerebral cortex show morphological and neurotransmitter heterogeneity during development", CEREB. CORTEX N. Y. N 1991, vol. 6, 1996, pages 490 - 497
LEAVITT, A.D.; ROBLES, G.; ALESANDRO, N.; VARMUS, H.E.: "Human immunodeficiency virus type 1 integrase mutants retain in vitro integrase activity yet fail to integrate viral DNA efficiently during infection", J. VIROL., vol. 70, 1996, pages 721 - 728, XP002207365
LERCHNER, W.; CORGIAT, B.; DER MINASSIAN, V.; SAUNDERS, R.C.; RICHMOND, B.J.: "Injection parameters and virus dependent choice of promoters to improve neuron targeting in the nonhuman primate brain", GENE THER, 2014
LIN, E.-J.D.; YOUNG, D.; BAER, K.; HERZOG, H.; DURING, M.J.: "Differential actions of NPY on seizure modulation via Y1 and Y2 receptors: evidence from receptor knockout mice", EPILEPSIA, vol. 47, 2006, pages 773 - 780, XP003018679, DOI: doi:10.1111/j.1528-1167.2006.00500.x
LOBB, C.: "Abnormal Bursting as a Pathophysiological Mechanism in Parkinson's Disease", BASAL GANGLIA, vol. 3, 2014, pages 187 - 195
LUNDBERG, C.; BJORKLUND, T.; CARLSSON, T.; JAKOBSSON, J.; HANTRAYE, P.; DEGLON, N.; KIRIK, D.: "Applications of lentiviral vectors for biology and gene therapy of neurological disorders", CURR. GENE THER., vol. 8, 2008, pages 461 - 473, XP009162891, DOI: doi:10.2174/156652308786847996
MAINARDI, M.; PIETRASANTA, M.; VANNINI, E.; ROSSETTO, O.; CALEO, M.: "Tetanus neurotoxin-induced epilepsy in mouse visual cortex", EPILEPSIA, vol. 53, 2012, pages e132 - 136
MCCOWN, T.J.: "Adeno-associated Virus-Mediated Expression and Constitutive Secretion of Galanin Suppresses Limbic Seizure Activity in Vivo", MOL THER, vol. 14, 2006, pages 63 - 68, XP005524562, DOI: doi:10.1016/j.ymthe.2006.04.004
NGUGI, A.K.; BOTTOMLEY, C.; KLEINSCHMIDT, I.; SANDER, J.W.; NEWTON, C.R.: "Estimation of the burden of active and life-time epilepsy: a meta-analytic approach", EPILEPSIA, vol. 51, 2010, pages 883 - 890
NIKITIDOU, L.; TORP, M.; FJORD-LARSEN, L.; KUSK, P.; WAHLBERG, L.U.; KOKAIA, M.: "Encapsulated galanin-producing cells attenuate focal epileptic seizures in the hippocampus", EPILEPSIA, vol. 55, 2014, pages 167 - 174
J.L. NOEBELS, M. AVOLI, M.A. ROGAWSKI, R.W. OLSEN, AND A.V. DELGADO-ESCUETA: "Jasper's Basic Mechanisms of the Epilepsies", 2012, NATIONAL CENTER FOR BIOTECHNOLOGY INFORMATION, article NOE', F.M.; SORENSEN, A.T.; KOKAIA, M.; VEZZANI, A.: "Gene therapy of focal onset epilepsy using adeno-associated virus vector-mediated overexpression of neuropeptide Y"
PALFI, S.; GURRUCHAGA, J.M.; RALPH, G.S.; LEPETIT, H.; LAVISSE, S.; BUTTERY, P.C.; WATTS, C.; MISKIN, J.; KELLEHER, M.; DEELEY, S.: "ong-term safety and tolerability of ProSavin, a lentiviral vector-based gene therapy for Parkinson's disease: a dose escalation, open-label, phase 1/2 trial", LANCET LOND. ENGL., vol. 383, 2014, pages 1138 - 1146, XP055161289, DOI: doi:10.1016/S0140-6736(13)61939-X
PICOT, M.-C.; BALDY-MOULINIER, M.; DAURES, J.-P.; DUJOLS, P.; CRESPEL, A.: "The prevalence of epilepsy and pharmacoresistant epilepsy in adults: a population-based study in a Western European country", EPILEPSIA, vol. 49, 2008, pages 1230 - 1238
RAHIM, A.A.; WONG, A.M.S.; HOWE, S.J.; BUCKLEY, S.M.K.; ACOSTA-SALTOS, A.D.; ELSTON, K.E.; WARD, N.J.; PHILPOTT, N.J.; COOPER, J.D: "Efficient gene delivery to the adult and fetal CNS using pseudotyped non-integrating lentiviral vectors", GENE THER., vol. 16, 2009, pages 509 - 520
RICHICHI, C.; LIN, E.-J.D.; STEFANIN, D.; COLELLA, D.; RAVIZZA, T.; GRIGNASCHI, G.; VEGLIANESE, P.; SPERK, G.; DURING, M.J.; VEZZA: "Anticonvulsant and antiepileptogenic effects mediated by adeno-associated virus vector neuropeptide Y expression in the rat hippocampus", J. NEUROSCI. OFF. J. SOC. NEUROSCI., vol. 24, 2004, pages 3051 - 3059, XP008068517, DOI: doi:10.1523/JNEUROSCI.4056-03.2004
SCHUELE, S.U.; LUDERS, H.O.: "Intractable epilepsy: management and therapeutic alternatives", LANCET NEUROL., vol. 7, 2008, pages 514 - 524
SNOWBALL, A.; SCHORGE, S.: "Changing channels in pain and epilepsy: Exploiting ion channel gene therapy for disorders of neuronal hyperexcitability", FEBS LETT., vol. 589, 2015, pages 1620 - 1634, XP029140791, DOI: doi:10.1016/j.febslet.2015.05.004
STREIT AK; DERST C; WEGNER S; HEINEMANN U; ZAHN RK; DECHER N: "RNA editing of Kv1.1 channels may account for reduced ictogenic potential of 4-aminopyridine in chronic epileptic rats", EPILEPSIA, vol. 52, 2011, pages 645 - 8, XP055492144, DOI: doi:10.1111/j.1528-1167.2011.02986.x
STREIT AK; MATSCHKE LA; DOLGA AM; RINNE S; DECHER N: "RNA editing in the central cavity as a mechanism to regulate surface expression of the voltage-gated potassium channel", J BIOL CHEM., vol. 289, 2014, pages 26762 - 71
SZYMCZAK, A.L.; VIGNALI, D.A.A.: "Development of 2A peptide-based strategies in the design of multicistronic vectors", EXPERT OPIN. BIOL. THER., vol. 5, 2005, pages 627 - 638, XP055378453, DOI: doi:10.1517/14712598.5.5.627
TOMLINSON, S.E.; RAJAKULENDRAN, S.; TAN, S.V.; GRAVES, T.D.; BAMIOU, D.E.; LABRUM, R.W.; BURKE, D.; SUE, C.M.; GIUNTI, P.; SCHORGE: "Clinical, genetic, neurophysiological and functional study of new mutations in episodic ataxia type 1", J NEUROL NEUROSURG PSYCHIATRY, 2013
WOLDBYE, D.P.D.; ANGEHAGEN, M.; GOTZSCHE, C.R.; ELBRAND-BEK, H.; SORENSEN, A.T.; CHRISTIANSEN, S.H.; OLESEN, M.V.; NIKITIDOU, L.;: "Adeno-associated viral vector-induced overexpression of neuropeptide Y Y2 receptors in the hippocampus suppresses seizures", BRAIN J. NEUROL., vol. 133, 2010, pages 2778 - 2788
WYKES, R.C., HEEROMA, J.H., KULLMANN, D.M., WALKER, M.C., AND SCHORGE, S.: "Lentiviral-mediated over-expression of the potassium channel Kv1.1 as a treatment for focal neocortical epilepsy", POSTER NO. 248.20/S6. 2011 NEUROSCIENCE MEETING PLANNER, - 2011
WYKES, R.C.; HEEROMA, J.H.; MANTOAN, L.; ZHENG, K.; MACDONALD, D.C.; DEISSEROTH, K.; HASHEMI, K.S.; WALKER, M.C.; SCHORGE, S.; KUL: "Optogenetic and potassium channel gene therapy in a rodent model of focal neocortical epilepsy", SCI. TRANSL. MED., vol. 4, 2012, pages 161ra152, XP055185925, DOI: doi:10.1126/scitranslmed.3004190
WYKES, R.C.; LIGNANI. G: "Gene therapy and editing: Novel potential treatments for neuronal channelopathies", NEUROPHARMACOLOGY, vol. 132, 2018, pages 108 - 117
YAGUCHI, M.; OHASHI, Y.; TSUBOTA, T.; SATO, A.; KOYANO, K.W.; WANG, N.; MIYASHITA, Y.: "Characterization of the properties of seven promoters in the motor cortex of rats and monkeys after lentiviral vector-mediated gene transfer", HUM. GENE THER. METHODS, vol. 24, 2013, pages 333 - 344
YANEZ-MUNOZ, R.J.; BALAGGAN, K.S.; MACNEIL, A.; HOWE, S.J.; SCHMIDT, M.; SMITH, A.J.; BUCH, P.; MACLAREN, R.E.; ANDERSON, P.N.; BA: "Effective gene therapy with nonintegrating lentiviral vectors", NAT. MED., vol. 12, 2006, pages 348 - 353, XP002433816, DOI: doi:10.1038/nm1365
Attorney, Agent or Firm:
KREMER, Simon et al. (GB)
Download PDF:
Claims:
Claims

1. An expression vector comprising an engineered KCNA 1 gene encoding an edited Kv1 .1 potassium channel operably linked to a promoter suitable to drive expression of the edited Kv1 .1 potassium channel in human cells,

wherein the engineered KCNA 1 gene has a nucleotide sequence having at least 90% sequence identity to the nucleotide sequence shown in SEQ ID NO: 1 , and

wherein the edited Kv1.1 potassium channel has an amino acid sequence having at least 90% sequence identity to the amino acid sequence shown in SEQ ID NO:2 and comprises a valine amino acid residue at a position corresponding to amino acid residue 400 shown in SEQ ID NO: 2.

2. An expression vector of claim 1 , wherein the edited Kv1.1 potassium channel has an amino acid sequence comprising or consisting the amino acid sequence shown in SEQ ID NO: 2.

3. An expression vector of claim 1 or claim 2, wherein the engineered KCNA 1 gene has a nucleotide sequence comprising or consisting of the nucleotide sequence shown in SEQ ID NO: 1 .

4. An expression vector of any one of claims 1-3, wherein the promoter is a cell type specific promoter.

5. An expression vector of claim 4, wherein the cell type specific promoter is specific for neurons.

6. An expression vector of claim 4 or claim 5, wherein the cell type specific promoter is specific for pyramidal neurons.

7. An expression vector of any of claims 1-6, wherein the cell type specific promoter comprises a human CAMK2A promoter. 8. An expression vector of claim 7, wherein the human CAMK2A promoter has a nucleotide sequence comprising or consisting of the nucleotide sequence shown in SEQ ID NO: 3 or a nucleotide sequence having at least 80% identity to the nucleotide sequence shown in SEQ ID NO: 3. ***

9. An expression vector of any one of claims 1-8 wherein the vector is a viral vector.

10. An expression vector of claim 9, wherein the viral vector is a lentiviral vector, optionally wherein the lentiviral vector is a non-integrating lentiviral vector.

1 1 . An expression vector of claim 10, wherein the lentiviral vector comprises a nucleotide sequence having at least 95% identity to the nucleotide sequence of SEQ ID NO: 9.

^ ***

12. An in vitro method of making viral particles comprising:

transducing mammalian cells with a lentiviral vector according to claim 10 or claim 1 1 and expressing viral packaging and envelope proteins necessary for particle formation0 in the cells; and

culturing the transduced cells in a culture medium, such that the cells produce lentiviral particles that are released into the medium.

13. An in vitro method of claim 12, wherein the method comprises transducing the5 mammalian cells with one or more viral packaging and envelope expression vectors that encode the viral packaging and envelope proteins necessary for particle formation.

14. An in vitro method of claim 12 or 13, wherein the one or more packaging proteins includes a non-functional integrase enzyme such that the lentiviral vector is unable to0 incorporate its viral genome into the genome of the cell.

15. An in vitro method of any one of claims 12-14 further comprising separating the viral particles from the culture medium and optionally concentrating the viral particles.^ ***

16. A viral particle produced by the method of any one of claims 12-15, the viral particle optionally comprising an RNA molecule transcribed from the expression vector of any of claims 9-1 1 .

0

17. A viral particle comprising a single stranded RNA molecule encoding an engineered KCNA 1 gene operably linked to a promoter suitable to drive expression in5 human cells,

wherein the engineered gene encodes an edited Kv1.1 potassium channel, wherein the engineered KCNA 1 gene has a nucleotide sequence having at least 90% sequence identity to the nucleotide sequence shown in SEQ ID NO: 1 and

wherein the edited Kv1.1 potassium channel has an amino acid sequence having0 at least 90% sequence identity to the amino acid sequence shown in SEQ ID NO:2 and comprises a valine amino acid residue at a position corresponding to amino acid residue 400 shown in SEQ ID NO: 2,

wherein the edited Kv1.1 potassium channel and engineered KCNA 1 gene are optionally as defined in claim 2 or claim 3,

5 wherein the promoter is optionally as defined in any one of claims 4 to 8, wherein the viral particle is optionally a lentiviral particle, which is optionally a non- integrating lentiviral particle.

18. A kit comprising an expression vector of claim 10 or 1 1 and one or more viral packaging and envelope expression vectors that encode viral packaging and envelope proteins necessary for particle formation when expressed in a cell. 19. A kit of claim 18, wherein the viral packaging expression vector is an integrase- deficient viral packaging expression vector.

20. A viral particle of claim 16 or claim 17 for use in a method of treatment of a human or animal body.

21 . A viral particle of claim 16 or claim 17 for use in a method of treatment of a neurological disorder.

22. A method of treatment of a neurological disorder comprising administering to an individual with the neurological disorder the viral particle of claim 16 or claim 17.

23. Use of a viral particle according to claim 16 or claim 17 in the manufacture of a medicament for the treatment of a neurological disorder.

24. A viral particle for use according to claim 21 , method of treatment of claim 22, or use of a viral particle according to claim 23, wherein the neurological disorder is associated with neuronal hyperexcitibility.

25. A viral particle for use according to claim 21 , method of treatment of claim 22, or use of a viral particle according to claim 23, wherein the neurological disorder is a seizure disorder. 26. A viral particle for use, method of treatment, or use of a viral particle according to claim 25, wherein the seizure disorder is epilepsy.

27. A viral particle for use, method of treatment, or use of a viral particle, according to claim 26 wherein the epilepsy is neocortical epilepsy.

28. A viral particle for use according to claim 21 , method of treatment of claim 22, or use of a viral particle according to claim 23, wherein the neurological disorder is

Parkinson's disease.

29. A viral particle for use according to claim 21 , method of treatment of claim 22, or use of a viral particle according to claim 23, wherein the neurological disorder is chronic pain.

^ ***

30. A method of confirming the presence of engineered KCNA 1 mRNA in a cell, the method comprising:

transducing a cell with an expression vector of any one of claims 1-12 or

0 administering a viral particle of claim 17 or claim 18 to a cell under conditions that permit expression of engineered KCNA 1 mRNA; and

detecting the presence of the engineered KCNA 1 mRNA in the cell using a hybridisation assay. 5 31 . An in vitro or ex vivo method of confirming the presence of engineered KCNA 1 mRNA in a cell that has been obtained from a subject administered with a viral particle of claim 17 or claim 18, the method comprising:

detecting the presence of engineered KCNA 1 mRNA in the cell using a hybridisation assay.

0

32. A method of claim 30 or claim 31 , wherein the hybridisation assay is an in situ hybridisation assay using a labelled RNA probe, optionally wherein the labelled RNA probe is fluorescently labelled.

^ ***

33. A cell comprising the expression vector of any one of claims 1-1 1.

34. A cell of claim 33, wherein the cell is a mammalian cell.

0

35. A cell of claim 34, wherein the cell is a human cell.

36. A cell of claim 35, wherein the cell is a human embryonic kidney cell. 5 ***

37. A nucleic acid comprising an engineered KCNA 1 gene encoding an edited Kv1.1 potassium channel, wherein the engineered KCNA 1 gene has a nucleotide sequence having at least 90% sequence identity to the nucleotide sequence shown in SEQ ID NO:0 1 and

wherein the edited Kv1.1 potassium channel has an amino acid sequence having at least 90% sequence identity to the amino acid sequence shown in SEQ ID NO:2 and comprises a valine amino acid residue at a position corresponding to amino acid residue 400 shown in SEQ ID NO: 2,

5 wherein the edited Kv1.1 potassium channel and engineered KCNA 1 gene are optionally as defined in claim 2 or claim 3.

Description:
Expression vectors comprising engineered genes

This application claims priority from GB1709551 .4 filed 15 June 2017, the contents and elements of which are herein incorporated by reference for all purposes.

Technical field

The present invention relates generally to methods and materials involving engineered genes encoding potassium channels which can be used in treating epilepsy and similar neurological disorders.

Background art

Epilepsy affects over 60 million people worldwide (Ngugi et al., 2010). Even with optimal treatment -30% remain resistant to pharmacotherapy (Kwan et al., 201 1 ; Picot et al., 2008). The development of new anti-epileptic drugs in the last 20 years has had little impact on refractory epilepsy; people with inadequately controlled seizures continue to experience major co-morbidities, social exclusion, and an annual rate of sudden unexpected death in epilepsy (SUDEP) of 0.5-1 % (Devinsky, 201 1 ; Hoppe and Elger, 201 1 ). Although surgery can result in seizure freedom for patients with refractory epilepsy, it is unsuitable in over 90% of cases. Neocortical focal epilepsy is particularly poorly suited to surgical intervention because of the high risk of damage to eloquent regions of the cortex involved in language, vision or fine motor control (Schuele and Lijders, 2008). People with focal neocortical epilepsy are often left with very few, usually palliative, treatment options. There is therefore an urgent need to develop new

treatments.

Gene therapy is one promising option (Kullmann et al., 2014), but major hurdles remain in achieving stable, predictable and safe transgene expression with viral vectors. Hitherto, clinical trials with lentivectors for CNS disorders have mainly been restricted to ex-vivo treatment of haematopoietic stem cells for enzyme defects (Biffi et al., 2013, 2013; Cartier et al., 2009); however a recent trial using a lentivector injected into the striatum has demonstrated safety and tolerability in Parkinson's disease, with some evidence of decreased L-DOPA requirement (Palfi et al., 2014).

Studies of gene therapy for epilepsy initially focused on acutely precipitated seizures, which often translate poorly (Galanopoulou et al., 2012). However, more recent strategies, mainly focusing on adeno-associated viruses (AAVs) in models of temporal lobe epilepsy, have shown that the development of seizures after an epileptogenic insult (epileptogenesis) can be attenuated (Bovolenta et al., 2010; Haberman et al., 2003;

Kanter-Schlifke et al., 2007; Lin et al., 2006; McCown, 2006; Noe' et al., 2012; Richichi et al., 2004; Woldbye et al., 2010).

Potassium ion channels normally reduce the propensity of neurons to fire and to release neurotransmitters. When introduced to the brain using viral vectors they are effective as tools to dampen brain excitability and to treat experimental epilepsy in rodents. Potassium channel gene therapy is therefore promising for the treatment of human epilepsy as well as other neuropsychiatric diseases where neurons fire excessively such as chronic pain. The association of potassium channels with neurological disorders, such as epilepsy, has been described (D'Adamo et al., 2013). Gene therapy strategies based on the up or downregulation of genes that modulate neuronal excitability have been described as a potential therapy (Wykes and Lignani, 2017). Previous approaches to gene therapy in a model of epilepsia partialis continua (EPC) induced by tetanus toxin (TeNT) injection into the rat motor cortex (Katzel et al., 2014; Wykes et al., 2012) have been described. In this model pathological high-frequency electrocorticographic (ECoG) activity is prominent, but discrete long-lasting seizures are rare. Lentiviral overexpression of the human potassium channel Kv1 .1 , encoded by KCNA 1, was highly effective at reducing pathological high frequency activity in the motor cortex (Wykes et al., 201 1 & 2012). In vitro studies showed that Kv1 .1 overexpression reduced both intrinsic neuronal excitability and glutamate release from transduced pyramidal neurons (Heeroma et al., 2009; Wykes et al., 2012). Importantly both effects were graded; that is, neither neuronal excitability nor neurotransmitter release was completely abolished. However, it remains unclear whether these graded effects on excitability and transmitter release, and the reduction of pathological ECoG activity in the motor cortex, can be extrapolated to commoner, longer-lasting focal seizures arising from other parts of the brain. Streit et al. (201 1 & 2014) describe that Kv1 .1 channels undergo enzymatic RNA deamination to generate a channel with a single amino acid exchange located in the inner pore cavity (Kv1.1 l400V ).

Therefore, there remains a need in the art for improved gene therapy tools for treating neurological diseases, such as epilepsy.

Disclosure of the invention

The present inventors have made a number of modifications to the KCNA 1 gene that unexpectedly can be combined to enhance the translation and activity of the encoded

Kv1 .1 protein and improve the detection of KCNA 1 gene expression in a cell. Briefly, the inventors have designed and tested an expression vector comprising an engineered KCNA 1 gene that encodes an edited potassium channel. As demonstrated herein, the engineered KCNA 1 gene of the present invention produces functional Kv1.1 channels and that when packaged in a lentiviral vector was able to significantly reduce seizure frequency when administered to rats in a randomised, blinded preclinical trial.

Aspects of the invention are defined in the claims appended hereto. In one aspect the invention provides an expression vector comprising an engineered

KCNA 1 gene encoding an edited Kv1 .1 potassium channel operably linked to a promoter suitable to drive expression of the edited Kv1 .1 potassium channel in human cells, wherein the edited Kv1.1 potassium channel comprises a valine amino acid residue at a position corresponding to amino acid residue 400 shown in SEQ ID NO: 2. In one embodiment the engineered KCNA 1 gene has a nucleotide sequence comprising or consisting of the nucleotide sequence shown in SEQ ID NO: 1.

In some embodiments, the expression vector is a viral vector, such as a lentiviral vector.

The invention also provides viral particles and in vitro methods of making viral particles as defined herein.

Further, the invention provides kits comprising viral vectors as defined herein and one or more viral packaging and envelope vectors as defined herein. Further, the invention provides a viral particle as defined herein for use in a method of treatment of the human or animal body.

Further, the invention provides methods of confirming the presence of engineered KCNA 1 RNA in a cell as defined herein and also an in vitro or ex vivo method of confirming the presence of engineered KCNA 1 RNA in a cell that has been obtained from a subject administered with a viral particle as defined herein.

The invention also provides cells comprising the expression vector as defined herein. Furthermore, the invention also provides a nucleic acid as defined herein.

Some particular aspects of the invention will now be discussed in more detail. Expression vectors

The invention provides an expression vector comprising an engineered KCNA 1 gene encoding an edited Kv1.1 potassium channel operably linked to a promoter as defined herein. An expression vector as used herein is a DNA molecule used to transfer and express foreign genetic material in a cell. Such vectors include a promoter sequence operably linked to the gene encoding the protein to be expressed. "Promoter" means a minimal DNA sequence sufficient to direct transcription of a DNA sequence to which it is operably linked. "Promoter" is also meant to encompass those promoter elements sufficient for promoter-dependent gene expression controllable for cell type specific expression; such elements may be located in the 5' or 3' regions of the native gene.

An expression vector may also include a termination codon and expression enhancers. Any suitable vectors, promoters, enhancers and termination codons may be used to express the edited Kv1.1 potassium channel from an expression vector according to the invention. Suitable vectors include plasmids, binary vectors, phages, phagemids, viral vectors and artificial chromosomes (e.g. yeast artificial chromosomes or bacterial artificial chromosomes). As described in more detail below, preferred expression vectors include viral vectors such as lentiviral vectors. An expression vector may additionally include a reporter gene encoding a reporter protein. An example of a reporter protein is a green fluorescent protein (GFP). A reporter gene may be operably linked to its own promoter or, more preferably, may be operably linked to the same promoter as the engineered KCNA 1 gene of the invention. As an example of the latter, the KCNA 1 gene and reporter gene may be located either side of a sequence encoding a 2A peptide, such as a T2A peptide. 2A peptides are short (-20 amino acids) sequences that permit multicistronic gene expression from single promoters by impairing peptide bond formation during ribosome-mediated translation (Szymczak and Vignali, 2005). Having the reporter gene operably linked to the same promoter as the engineered KCNA 1 gene is thought to act as a reliable indicator of KCNA 1 gene expression. An expression vector including a reporter gene may be particularly useful in for preclinical applications, for example for use in animal models where it would be it can be used to help assess the localisation of gene expression.

In other embodiments, the expression vector lacks a sequence encoding a reporter protein. This may be preferred for regulatory reasons, for example. In embodiments of the invention, reporting or detecting the KCNA 1 gene of the invention may be achieved in different ways - for example based on its engineered sequence.

Generally speaking, those skilled in the art are well able to construct vectors and design protocols for recombinant gene expression. Suitable vectors can be chosen or constructed, containing, in addition to the elements of the invention described above, appropriate regulatory sequences, including promoter sequences, terminator fragments, polyadenylation sequences, marker genes and other sequences as appropriate.

Molecular biology techniques suitable for the expression of polypeptides in cells are well known in the art. For further details see, for example, Molecular Cloning: a Laboratory Manual: 2nd edition, Sambrook et al, 1989, Cold Spring Harbor Laboratory Press or Current Protocols in Molecular Biology, Second Edition, Ausubel et al. eds., John Wiley & Sons, (1995, and periodic supplements). The term "operably linked" used herein includes the situation where a selected gene and promoter are covalently linked in such a way as to place the expression of the gene (i.e. polypeptide coding) under the influence or control of the promoter. Thus, a promoter is operably linked to an engineered KNCA 1 gene if the promoter is capable of effecting transcription of the gene into RNA in a cell. Where appropriate, the resulting RNA transcript may then be translated into a desired protein or polypeptide. The promoter is suitable to effect expression of the operably linked gene in a mammalian cell. Preferably, the mammalian cell is a human cell.

KCNA 1 qenes and Kv1.1 potassium channels KCNA 1 (Gene ID 3736, also known as the Potassium Voltage-Gated Channel Subfamily A Member 1, KV1. 1, HBK1 and RBK1) is a human gene that encodes the human Kv1.1 potassium channel (also known as Potassium voltage-gated channel subfamily A member 1 ). By "wild-type KCNA 1 gene" it is meant the nucleic acid molecule that is found in human cells and encodes the human Kv1 .1 potassium channel. The KCNA 1 gene may include regulatory sequences upstream or downstream of the coding sequence. A nucleotide sequence for the wild-type KCNA 1 gene, including the non- coding 5' and 3' untranslated regions (5' and 3' UTRs) is provided in NCBI Reference Sequence NM_000217.2. The coding sequence for the wild-type KCNA 1 gene has the nucleotide sequence of SEQ ID NO: 4, which corresponds to positions 1 106 to 2593 of NCBI Reference Sequence NM_000217.2.

The Kv1 .1 potassium channel is a voltage-gated delayed potassium channel that is phylogenetically related to the Drosophila Shaker channel. The amino acid sequence for the wild-type Kv1 .1 potassium channel has the amino acid sequence of SEQ ID NO: 5 which is identical to the NCBI Reference Sequence NP_000208.2. Voltage-dependent potassium channels modulate excitability by opening and closing a potassium-selective pore in response to voltage. In many cases, potassium ion flow can be interrupted when an intracellular particle occludes the pore, a process known as fast inactivation. The Kv1 .1 potassium channel has six putative transmembrane segments, and the loop between the fifth and sixth segment forms the pore.

During normal production in cells, some of the KCNA 1 RNA in the cell is edited by an adenosine deaminase acting on RNA (ADAR) that causes an isoleucine/valine (l/V) recoding event at a single position I400 that lies within the sixth transmembrane domain and lines the inner vestibule of the ion-conducting pore (Hoopengardner et al., Science 301 (5634):832-6, 2003). At negative membrane potentials, channels containing unedited I400 recover from inactivation at a rate around twenty times slower than their edited (V400) counterparts (Bhalla et al., 2004).

The present invention involves the expression of an edited Kv1 .1 potassium channel.

An "edited Kv1 .1 potassium channel" is a functional Kv1.1 potassium channel but contains the isoleucine/valine mutation described above. Without wishing to be bound by any particular theory, it is believed that providing an edited Kv1 .1 potassium channel is advantageous as it immediately provides the cell with edited Kv1.1 potassium channels, without relying on the ADAR-dependent RNA edited step that normally occurs in the cell. It is believed that these edited Kv1 .1 potassium channels are much quicker at recovering from inactivation than their unedited counterparts.

Furthermore, not all of the KCNA 1 RNA is normally edited in the cell. As described by Hoopengardner et al., Science 301 (5634):832-6, 2003 (see, in particular, Fig. 3) the amount of RNA editing that occurs varies across different regions of the human nervous system, ranging from around 17% in the caudate nucleus to around 77% in the medulla. For example, around 20% of KCNA 1 RNA is believed to be edited in the frontal cortex meaning that around 80% of KCNA 1 RNA remains unedited in this region of the brain. As shown in the examples herein, providing an engineered KCNA 1 gene that encodes an edited Kv1 .1 channel was able to efficiently reduce seizure frequency when administered to rats in a randomised, blinded, preclinical trial. As used herein, an edited Kv1.1 potassium channel has an amino acid sequence having at least 70, 75, 80, 85, 90, 91 , 92, 93, 94, 95, 96, 97, 98, 99 or 100% sequence identity to the amino acid sequence shown in SEQ ID NO: 2 provided it also contains a valine amino acid residue at a position corresponding to amino acid residue 400 shown in SEQ ID NO: 2 (the 'edited position'). In some preferred embodiments, the edited Kv1 .1 potassium channel has an amino acid sequence comprising or consisting of the amino acid sequence shown in SEQ ID NO: 2.

An edited Kv1.1 potassium channel that contains a valine amino acid residue at a position corresponding to amino acid residue 400 shown in SEQ ID NO: 2 can be identified by methods known in the art. For example, the edited position can be identified by a sequence alignment between the amino acid sequence of SEQ ID NO: 2 and the amino acid sequence of the edited Kv1.1 potassium channel of interest. Such sequence alignments can then be used to identify the edited position in the edited Kv1.1 potassium channel of interest which, at least in the alignment, is near, or at the same position as, the edited position at amino acid residue 400 in the amino acid sequence shown in SEQ ID NO: 2.

A functional Kv1 .1 potassium channel is a protein that retains the normal activity of a potassium channel, e.g. the channels are able to open and close in response to voltage. Methods of testing that the Kv1 .1 potassium channels are functional are known in the art and some of which are described herein. Briefly, a suitable method for confirming that the Kv1.1 potassium channel is functional involves transfecting cells with an expression vector encoding a Kv1.1 potassium channel and using electrophysiological techniques such as patch clamping to record currents of the potassium channels.

The wild-type Kv1.1 potassium channel comprises a tyrosine amino acid at position 379 as shown in SEQ ID NO: 5. In some embodiments, an edited Kv1 .1 potassium channel comprises a tyrosine amino acid residue at a position corresponding to amino acid residue 379 shown in SEQ ID NO: 2.

In other embodiments, an edited Kv1 .1 potassium channel comprises a valine amino acid residue at a position corresponding to amino acid residue 379 shown in SEQ ID NO: 2. An example of an edited Kv1 .1 potassium channel with this amino acid sequence is shown in SEQ ID NO: 14. Without wishing to be bound by any particular theory, it is believed that a Y379V mutation reduces the sensitivity of Kv1.1 channels to

tetraethylammonium (TEA) without altering the functional properties of the potassium channel. For example, this change in sensitivity allows transgenic Kv1.1 channels to be pharmacologically isolated from their wild-type counterparts in patch clamp

electrophysiology experiments (Heeroma et al. 2009). In the present invention an "engineered KCNA 1 gene" is used. An engineered KCNA 1 gene differs from the nucleotide sequence of the wild-type KCNA 1 gene as described herein but still encodes for a functional Kv1 .1 potassium channel. As used herein, an engineered KCNA 1 gene has a nucleotide sequence having at least 70, 75, 80, 85, 90, 91 , 92, 93, 94, 95, 96, 97, 98, 99 or 100% sequence identity to the nucleotide sequence shown in SEQ ID NO: 1. In some preferred embodiments, the engineered KCNA 1 gene has a nucleotide sequence comprising or consisting of the nucleotide sequence shown in SEQ ID NO: 1 . As described above, an embodiment of the invention includes an engineered KCNA 1 gene encoding an edited potassium channel that comprises a valine amino acid residue at position 379, as shown in SEQ ID NO: 14. An example of an engineered KCNA 1 gene that encodes the amino acid sequence shown in SEQ ID NO: 14 is the nucleotide sequence shown in SEQ ID NO: 13. In some embodiments, the engineered KCNA 1 gene has a nucleotide sequence comprising or consisting of the nucleotide sequence shown in SEQ ID NO: 13, or has at least 75, 80, 85, 90, 91 , 92, 93, 94, 95, 96, 97, 98, 99 or 100% sequence identity to the nucleotide sequence shown in SEQ ID NO: 13.

Cell type specific promoters

Previous studies relied on driving overexpression of KCNA 1 with a strong viral promoter (CMV), which in rat demonstrated a strong bias for excitatory neurons (Wykes et al., 2013). However, there are safety concerns associated with the use of non-specific promoters such as the CMV promoter. For example, in the case of epilepsy, a concern is the possibility of potassium channel overexpression in cells other than excitatory neurons, for example interneurons, which could aggravate seizure activity by exacerbating rather than attenuating local excitability. A method of improving safety would therefore be to restrict expression of the Kv1 .1 potassium channel to particular cell types. However it is not clear if promoters that drive cell type specificity would provide sufficient expression to have a functional effect in vivo, particularly in the case where an edited Kv1.1 potassium channel is expressed.

The present inventors found that the improved expression as a result of the engineered KCNA 1 gene meant that a cell type specific promoter could be used to drive expression of this gene.

Thus, in some embodiments, the promoter is a cell type specific promoter that drives cell type specific expression of the edited Kv1 .1 potassium channel. A cell type specific promoter is a promoter that drives more expression in the cell type of interest than in other cell types. For example, where the cell type specific promoter is specific for neurons, this will drive more expression in neurons than in other cell types, for example glial cells. Preferably, the cell type specific promoter means at least 60%, 70%, 80%, 90%, 95%, 96%, 97%, 98% 99% or most preferably 100% of expression occurs in the cell type of interest than in another cell type. Methods of determining gene and protein expression and localisation are known in the art. They include assays that detect RNA transcripts, such as hybridisation methods described herein, as well as and methods that detect protein such as

immunohistochemical methods. One such method for assessing the cell type specificity of the cell type specific promoter would be to compare the overlap of RNA transcripts or protein with markers for particular cell types. For example, where the cell type specific promoter is believed to be specific for neurons, localisation of the RNA or protein for the gene of interest operably linked to this promoter can be compared for overlap with localisation of known immunohistochemical markers for neurons, e.g. NeuN, and glial cells, e.g. GFAP. The promoter will be considered a neuron cell type specific promoter if greater overlap is observed between the gene of interest and neurons than between the gene of interest and glial cells.

The cell type specific promoter that is used will depend on the cell type that is being targeted. For example, in the case of a treating a neurological disorder, it may be preferable to target neural cells, such as neurons and glial cells. In particularly preferred examples, the cell type specific promoters is specific for neurons, in other words it drives higher levels of expression in neurons than in glial cells. In some cases, the cell type specific promoter is specific for excitatory neurons, such as glutamatergic neurons. An example of an excitatory neuron is a pyramidal neuron. Glutamatergic neurons can be identified by detecting markers that are specific for gluatamatergic cells, such as vGlutl , vGlut2, NMDAR1 , NMDAR2B, glutaminase, glutamine synthetase. Without wishing to be bound by any particular theory, cell type specific expression of the engineered KCNA 1 gene in glutamatergic neurons is believed to be useful for treatment of diseases associated with neuronal hyperexcitability, in particular epilepsy.

A preferred example of the neuronal cell type specific promoter is the human CAMK2A (alpha CaM kinase II gene) promoter. The CAMK2A promoter is known to bias expression to excitatory neurons and furthermore leads to very little expression in GABAergic cells (also known as interneurons) (Dittgen et al., 2004; Yaguchi et al., 2013). The CAMK2A promoter is therefore an example of a cell type specific promoter that is specific for excitatory neurons. The CAMK2A promoter may have a nucleotide sequence comprising or consisting of the nucleotide sequence of SEQ ID NO: 3. Alternatively, the CAMK2A promoter may have a nucleotide sequence comprising or consisting of a nucleotide sequence having at least 70, 75, 80, 85, 90, 91 , 92, 93, 94, 95, 96, 97, 98 or 99% identity to the nucleotide sequence of SEQ ID NO: 3.

Another promoter that is believed to be specific for neurons is the VGLUT1 promoter (Zhang et al. Brain Research 1377:1 -12, 201 1 , herein incorporated by reference at least for the sequence of the promoters and related sequences). As described by Zhang et al., the rat VGLUT1 upstream promoter or the first intron, after fusion to a basal promoter, results in glutamatergic-specific expression. A further example of a promoter that has been shown to be specific for glutamatergic neurons in rats is the PAG promoter

(Rasmussen et al. Brain Research 1 144: 19-32, 2007, herein incorporated by reference at least for the sequence of the promoters and related sequences). Other neuronal cell type-specific promoters include the NSE promoter (Liu H. et al., Journal of Neuroscience. 23(18):7143-54, 2003 & Peel AL. et al., Gene Therapy. 4(1 ): 16- 24, 1997); tyrosine hydroxylase promoter (Kessler MA. et al., Brain Research. Molecular Brain Research. 1 12(l-2):8-23, 2003); myelin basic protein promoter (Kessler MA. et al Biochemical & Biophysical Research Communications. 288(4):809-18, 2001 );

neurofilaments gene (heavy, medium, light) promoters (Yaworsky PJ.et al., Journal of Biological Chemistry. 272(40):251 12-20, 1997) (All of which are herein incorporated by reference at least for the sequence of the promoters and related sequences.). A further suitable promoter is the Synapsinl promoter (see KQgler et al "Human synapsin 1 gene promoter confers highly neuron-specific long-term transgene expression from an adenoviral vector in the adult rat brain depending on the transduced area." Gene

Therapy. 10(4):337-47 2003).

The Nav1 .7 and Nav1 .8 sodium channels are thought to play an important role in pain and therefore their promoter could be used for the treatment of diseases such as chronic pain. The human gene that encodes Nav1 .7 is SCN9A and the identification and use of the SCN9A promoter has been described, for example, in Diss et al. Molecular and Cellular Neuroscience. 37(3): 537-47, 2008. The human gene that encodes Nav1.8 is SCN10A and the identification and use of the SCN10A has been described, for example, in Puhl & Ikeda, Journal of Neurochemistry. 106(3): 1209-24, 2008. These references are herein incorporated by reference at least for the sequence of the promoters and related sequences.

Viral vectors

A preferred expression vector for use with the present invention is a viral vector, such as a lentiviral or AAV vector. A particularly preferred expression vector is a lentiviral vector. Lentiviral vectors are a special type of retroviral vector which, when assembled into viral particles, are typically characterized by having a long incubation period for infection and are able to infect non-dividing cells, such as post-mitotic neurons. Lentiviral vectors generally lead to rapid, stable and spatially-restricted expression (Lundberg et al., 2008). This may be optimal for treatment of disease such as epilepsy where focal seizures often arise from brain areas very close to eloquent cortex. In addition, the large packaging capacity of lentivectors allows a greater choice of promoter-transgene combinations (Kantor et al., 2014), which can further increase the specificity of expression.

Lentiviral vectors are based on the nucleic acid backbone of a virus from the lentiviral family of viruses. The infectious process of a lentivirus found in nature is well known to the skilled person. Without wishing to be bound by any particular theory, a brief description of this is provided here. A lentivirus is a retrovirus, meaning it has a single stranded RNA genome and a reverse transcriptase enzyme. This single stranded RNA genome is packaged within a viral envelope with protruding glycoproteins that aid in attachment to a host cell's outer membrane. Within the viral genome are nucleic acid sequences, including the gag, po/ and env gene regions. The po/ gene encodes the reverse transcriptase enzyme, required for reverse transcription of the viral RNA genome and the integrase enzyme, required for efficient integration of the viral genome into the host cell genome. The env gene encodes various envelope proteins and the gag gene encodes various structural proteins.

During infection, the viral material is injected into the host cell where the viral reverse transcriptase performs reverse transcription of the viral RNA genome to create a viral DNA genome. The viral DNA is then incorporated into the host cell's genome. From there the host cell performs transcription and translation to create viral particles which burst from the host cell and can go on to infect other host cells. Lentiviral vectors have been developed by removing the non-essential sequences and genomic regions involved with viral replication and virulence from the wild-type lentiviral genome, resulting in a replication defective vector containing the necessary elements for packaging and processing (Shaw & Cornetta, Biomedicines 2(1 ): 14-35, 2014). As used herein a lentiviral vector means a DNA expression vector which comprises the lentiviral genes sufficient to result in the engineered KCNA 1 gene being transcribed into RNA and which RNA is packaged into a viral particle when expressed alongside lentiviral envelope and packaging proteins. Typically, the lentiviral vector contains 5' and 3' long terminal repeat (LTR) regions of a lentivirus, such as SIV and HIV. The 5' LTR can act as an RNA polymerase promoter. In some lentiviral vectors, part of the 5' LTR promoter, such as the U3 region, is replaced by another promoter, generally a constitutive promoter such as a CMV or RSV promoter. The lentiviral vector may also contain a bacterial plasmid portion, additional lentiviral elements required for viral vector RNA packaging and intracellular transport, a marker gene and elements for their regulation, optional chromatin-control elements and sites for convenient plasmid DNA re-engineering.

In preferred embodiments, the lentiviral vector comprises, in order from 5' to 3', a CMV enhancer/promoter, a truncated 5' LTR, a HIV-1 packaging signal, a Rev response element, a central polypurine tract and central termination sequence (cPPT/CTS), a cell type specific promoter (such as a human CAMK2A promoter), an engineered KCNA 1 gene, a woodchuck hepatitis virus post-translational regulatory element and a 3' LTR. In some embodiments, the lentiviral vector contains a sequence encoding a reporter protein, such as a fluorescent protein. An example of a lentiviral vector containing a sequence encoding GFP is shown in SEQ ID NO: 7. In other embodiments the lentiviral vector lacks a sequence encoding a reporter protein, such as a fluorescent protein.

In some embodiments, the lentiviral vector comprises a nucleotide sequence having at least 70, 75, 80, 85, 90, 91 , 92, 93, 94, 95, 96, 97, 98 or 99% identity to the nucleotide sequence of SEQ ID NO: 9 or SEQ ID NO: 1 1. In some embodiments, the lentiviral vector comprises the nucleotide sequence of SEQ ID NO: 9 or SEQ ID NO: 1 1. SEQ ID NO: 9 encodes an edited Kv1 .1 potassium channel with a tyrosine at position 379 whilst SEQ ID NO: 1 1 encodes an edited Kv1.1 potassium channel with a valine at position 379.

In some embodiments, the lentiviral vector comprises a nucleotide sequence having at least 70, 75, 80, 85, 90, 91 , 92, 93, 94, 95, 96, 97, 98 or 99% identity to the nucleotide sequence of SEQ ID NO: 10 or SEQ ID NO: 12. In some embodiments, the lentiviral vector comprises or consists of the nucleotide sequence of SEQ ID NO: 10 or SEQ ID NO: 12. SEQ ID NO: 10 encodes an edited Kv1 .1 potassium channel with a tyrosine at position 379 whilst SEQ ID NO: 12 encodes an edited Kv1.1 potassium channel with a valine at position 379.

In some embodiments, the lentiviral vector additionally comprises genes encoding viral packaging and envelope proteins.

In some embodiments, the lentiviral vector is a non-integrating lentiviral vector (NILV). Vector particles produced from these vectors do not integrate their viral genome into the genome of the cells and therefore are useful in applications where transient expression is required or for sustained episomal expression such as in quiescent cells. NILVs can be developed by mutations in the integrase enzyme or by altering the 5' LTR and/or the 3' LTR to prevent integrase from attaching these sequences. These modifications eliminate integrase activity without affecting reverse transcription and transport of the pre- integration complex to the nucleus. Without wishing to be bound by any particular theory, when a NILV enters a cell the lentiviral DNA is expected to remain as remains in the nucleus as an episome, leading to sustained expression in cell, e.g. post-mitotic cells. As demonstrated herein, the combined use of a NILV and a cell type specific promoter such as CAMK2A means that expression can be effectively targeted to particular cell types such as neurons.

Methods of generating NILVs are described by Shaw & Cornetta Biomedicines 2^ ): 14- 35, 2014, which is herein incorporated by reference. Examples of a mutation that is used to inhibit integration is the D64 mutation in an integrase enzyme, for example a D64V substitution. This and other examples of suitable modifications that inhibit integration are described in, e.g. Table 1 of Shaw & Cornetta, Biomedicines 2(1 ): 14-35, 2014.

Viral particles

The invention also includes in vitro methods of making lentiviral particles. In one embodiment, this method involves transducing mammalian cells with a lentiviral vector as described herein and expressing viral packaging and envelope proteins necessary for particle formation in the cells and culturing the transduced cells in a culture medium, such that the cells produce lentiviral particles that are released into the medium. An example of a suitable mammalian cell is a human embryonic kidney (HEK) 293 cell.

It is possible to use a single expression vector that encodes all the lentiviral components required for viral particle formation and function. Most often, however, multiple plasmid expression vectors or individual expression cassettes integrated stably into a host cell are utilised to separate the various genetic components that generate the lentiviral vector particles.

In some embodiments, expression cassettes encoding the one or more viral packaging and envelope proteins have been integrated stably into a mammalian cell. In these embodiments, transducing these cells with a lentiviral vector described herein is sufficient to result in the production of lentiviral particles without the addition of further expression vectors.

In other embodiments, the in vitro methods involve using multiple expression vectors. In some embodiments, the method comprises transducing the mammalian cells with one or more expression vectors encoding the viral packaging and envelope proteins that encode the viral packaging and envelope proteins necessary for particle formation.

Examples of suitable viral packaging and envelope proteins and expression vectors encoding these proteins are commercially available and well known in the art. In general, the viral packaging expression vector or expression cassette expresses the gag, pol, rev, and tat gene regions of HIV-1 which encode proteins required for vector particle formation and vector processing. In general, the viral envelope expression vector or expression cassette expresses an envelope protein such as VSV-G. In some cases, the packaging proteins are provided on two separate vectors - one encoding Rev and one encoding Gag and Pol. Examples of lentiviral vectors along with their associated packaging and envelope vectors include those of Dull, T. et al., "A Third-generation lentivirus vector with a conditional packaging system" J. Wro/ 72(1 1 ):8463-71 (1998), which is herein incorporated by reference.

An example of a viral envelope expression vector is pMDG-VSV-G, which contains the vsv-g gene operably linked to a CMV promoter. The construction of this vector is described by Kafri et al. 1999 J Virol. 73(1 ): 576-584, which is herein incorporated by reference.

An example of a viral packaging expression vector is pCMVdR8.74, which contains the gag, pol, tat and rev genes operably linked to a CMV promoter. This viral packaging expression vector is available, for example, from Addgene (Cambridge, MA, USA) as plasmid number 22036.

As explained above, in some embodiments the viral packaging expression vector is an integrase-deficient viral packaging expression vector. For example, the integrase- deficient viral packaging expression vector may encode a non-functional (e.g. mutated) integrase enzyme, such as a mutant D64V integrase. Vector particles produced from these vectors having non-functional integrases do not efficiently integrate their viral genome into the genome of the cells. An example of an integrase-deficient viral packaging expression vector is pCMVdR8.74 D64V , which contains the gag, pol, faf and rev genes operably linked to a CMV promoter and where the pol gene encodes a mutant D64V integrase. pCMVdR8.74 D64V is a 2 nd generation human immunodeficiency virus 1 (HIV-1 ) packaging plasmid with an aspartic acid 64 -valine mutation in its integrase coding sequence. This mutation reduces genomic integration by a factor of 10,000 compared to wild-type levels (Leavitt et al. 1996). The construction and use of this vector is described in Leavitt et al. 1996 and Yafiez-Mufioz et al. Nature Medicine. 12(3): 348-53, 2006, which are herein incorporated by reference. Following release of viral particles, the culture medium comprising the viral particles may be collected and, optionally the viral particles may be separated from the culture medium. Optionally, the viral particles may be concentrated.

Following production and optional concentration, the viral particles may be stored, for example by freezing at -80°C ready for use by administering to a cell and/or use in therapy.

The invention also provides viral particles, for example those produced by the methods described herein. As used herein, a viral particle comprises a RNA genome packaged within the viral envelope that is capable of infecting a cell, e.g. a mammalian cell. A viral particle may be integrase deficient, e.g. it may contain a mutant integrase enzyme or contain alterations in the 5' and/or 3' LTRs as described herein.

Kits

The invention also provides kits that comprise a lentiviral vector as described herein and one or more packaging and envelope expression vectors also described herein. In some embodiments the viral packaging expression vector is an integrase-deficient viral packaging expression vector, such as the pCMVdR8.74 D64V vector described herein.

Disorders

The viral vectors and viral particles described herein may also be for use as a

medicament. For example, a viral particle as described herein may be used in gene therapy.

The invention provides the use of a viral particle as described herein for the manufacture of a medicament for the treatment of a human or animal body, a viral particle as described herein for use in the treatment of a human or animal body and a method of treatment which comprises administering the viral particle as described herein to an individual in need thereof.

In certain embodiments, the viral particle is used in the treatment of a neurological disorder. Non-limiting examples of neurological disorders include seizure disorders (such as epilepsy), Alzheimer's disease, multiple sclerosis, Parkinson's disease, tremor and other movement disorders, chronic pain, migraine, and other neuropsychiatric diseases associated with alterations in neuronal excitability, including major depression, bipolar disorder, anxiety, and schizophrenia.

In certain embodiments, the neurological disorder is associated with neuronal

hyperexcitibility. Non-limiting examples of neurological disorders associated with neuronal hyperexcitability include seizure disorders (such as epilepsy), Alzheimer's disease, multiple sclerosis, Parkinson's disease, tremor and other movement disorders, chronic pain, migraine, major depression, bipolar disorder, anxiety, and schizophrenia. In preferred embodiments the treatment is for epilepsy, chronic pain, depression or Parkinson's disease.

In particularly preferred embodiments, the treatment is for epilepsy, for example idiopathic, symptomatic and cryptogenic epilepsy. The epilepsy may be neocortical epilepsy. The treatments described herein may be used to quench or blocking epileptogenic activity. The treatments may be used to reduce the frequency of seizures. The treatments may be used to temporally or permanently reduce neuronal excitability in the neurons.

Administration and dosage

The viral particles described herein can be delivered to the subject in a variety of ways, such as direct injection of the viral particles into the brain. For example, the treatment may involve direct injection of the viral particles into the cerebral cortex, in particular the neocortex. The treatment may involve direct injection of the viral particles into the location in the brain where it is believed to be functionally associated with the disorder. For example, where the treatment is for epilepsy this may involve direct injection of the viral particles into the motor cortex; where the treatment is for chronic pain, this may involve direct injection of the viral particles into the dorsal root ganglion; and where the treatment is for Parkinson's disease, this may involve direct injection of the viral particles into the Substantia Nigra. The particular method and site of administration would be at the discretion of the physician who would also select administration techniques using his/her common general knowledge and those techniques known to a skilled practitioner.

The invention may also be used to treat multiple epileptic foci simultaneously by injection directly into the multiple identified loci.

The patient may be one who has been diagnosed as having drug-resistant or medically- refractory epilepsy, by which is meant that epileptic seizures continue despite adequate administration of antiepileptic drugs.

The subject may be one who has been diagnosed as having well defined focal epilepsy affecting a single area of the neocortex of the brain. Focal epilepsy can arise, for example, from developmental abnormalities or following strokes, tumours, penetrating brain injuries or infections.

Following administration of the viral particles, the recipient individual may exhibit reduction in symptoms of the disease or disorder being treated. For example, for an individual being treated who has a seizure disorder such as epilepsy, the recipient individual may exhibit a reduction in the number of seizures. This may have a beneficial effect on the disease condition in the individual.

The term "treatment," as used herein in the context of treating a condition, pertains generally to treatment and therapy of a human, in which some desired therapeutic effect is achieved, for example, the inhibition of the progress of the condition, and includes a reduction in the rate of progress, a halt in the rate of progress, regression of the condition, amelioration of the condition, and cure of the condition. Treatment as a prophylactic measure (i.e., prophylaxis, prevention) is also included. The viral particle can be delivered in a therapeutically-effective amount.

The term "therapeutically-effective amount" as used herein, pertains to that amount of the viral particle which is effective for producing some desired therapeutic effect,

commensurate with a reasonable benefit/risk ratio, when administered in accordance with a desired treatment regimen.

Similarly, the term "prophylactically effective amount," as used herein pertains to that amount of the viral particle which is effective for producing some desired prophylactic effect, commensurate with a reasonable benefit/risk ratio, when administered in accordance with a desired treatment regimen.

"Prophylaxis" in the context of the present specification should not be understood to circumscribe complete success i.e. complete protection or complete prevention. Rather prophylaxis in the present context refers to a measure which is administered in advance of detection of a symptomatic condition with the aim of preserving health by helping to delay, mitigate or avoid that particular condition.

While it is possible for the viral particle to be used (e.g., administered) alone, it is often preferable to present it as a composition or formulation e.g. with a pharmaceutically acceptable carrier or diluent.

The term "pharmaceutically acceptable," as used herein, pertains to compounds, ingredients, materials, compositions, dosage forms, etc., which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of the subject in question (e.g., human) without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio. Each carrier, diluent, excipient, etc. must also be "acceptable" in the sense of being compatible with the other ingredients of the formulation. In some embodiments, the composition is a pharmaceutical composition (e.g., formulation, preparation, medicament) comprising, or consisting essentially of, or consisting of as a sole active ingredient, viral particle as described herein, and a pharmaceutically acceptable carrier, diluent, or excipient. As described in WO2008096268, in gene therapy embodiments employing delivery of the viral particle, the unit dose may be calculated in terms of the dose of viral particles being administered. Viral doses include a particular number of virus particles or plaque forming units (pfu). For embodiments involving adenovirus, particular unit doses include 10 3 , 10 4 , 10 5 , 10 6 , 10 7 , 10 8 , 10 9 , 10 10 , 10 11 , 10 12 , 10 13 or 10 14 pfu. Particle doses may be somewhat higher (10 to 100 fold) due to the presence of infection-defective particles. In some embodiments the methods or treatments of the present invention may be combined with other therapies, whether symptomatic or disease modifying.

The term "treatment" includes combination treatments and therapies, in which two or more treatments or therapies are combined, for example, sequentially or simultaneously.

For example it may be beneficial to combine treatment with a compound as described herein with one or more other (e.g., 1 , 2, 3, 4) agents or therapies. Appropriate examples of co-therapeutics will be known to those skilled in the art on the basis of the disclosure herein. Typically the co-therapeutic may be any known in the art which it is believed may give therapeutic effect in treating the diseases described herein, subject to the diagnosis of the individual being treated. For example epilepsy can sometimes be ameliorated by directly treating the underlying etiology, but anticonvulsant drugs, such as phenytoin, gabapentin, lamotrigine, levetiracetam, carbamazepine and clobazam, and topiramate, and others, which suppress the abnormal electrical discharges and seizures, are the mainstay of conventional treatment (Rho & Sankar, 1999, Epilepsia 40: 1471-1483). The particular combination would be at the discretion of the physician who would also select dosages using his/her common general knowledge and dosing regimens known to a skilled practitioner.

The agents (i.e. viral particle, plus one or more other agents) may be administered simultaneously or sequentially, and may be administered in individually varying dose schedules and via different routes. For example, when administered sequentially, the agents can be administered at closely spaced intervals (e.g., over a period of 5-10 minutes) or at longer intervals (e.g., 1 , 2, 3, 4 or more hours apart, or even longer periods apart where required), the precise dosage regimen being commensurate with the properties of the therapeutic agent(s).

Methods of confirming presence of KCNA 1

The invention also provides a method of confirming the presence of engineered KCNA 1 in a cell.

Overexpressing a normal human brain potassium channel avoids the risk of immune reactions, which potentially limit the clinical translation of gene therapy using other membrane proteins. However, a limitation of clinical translation using the normal gene sequence of human potassium channels is that it is difficult to detect their expression against the background endogenous channels present in the brain.

Because the sequence of the engineered KCNA 1 gene differs from the wild-type KCNA 1 gene found in cells, when this gene is transcribed into RNA it incorporates a unique RNA sequence (an 'RNA-fingerprint'). This RNA-fingerprint permits specific tracking of transgene expression with RNA-targeted techniques that would otherwise fail to distinguish between transgenic and endogenous KCNA 1. This is particularly useful where it is important to determine the localisation of engineered KCNA 1 gene expression without having to include sequences encoding fluorescent tags or epitopes that could potentially result in immunogenicity.

For example, tissue removed from patients who have been treated with the engineered KCNA 1 gene could be examined to determine where and in which cell types (excitatory neurons as expected, or inhibitory neurons or glial cells) the KCNA1 RNA was present. Such tissue could be obtained, for instance, from epilepsy surgery in the event of epilepsy gene therapy failure, or post-mortem. This data is expected to be useful for preclinical dosage calculation, biodistribution studies, regulatory approval and further clinical development on potassium channel gene therapy.

Thus, in one embodiment the method comprises transducing a cell with an expression vector as described herein or administering a viral particle as described herein to a cell under conditions that permit expression of engineered KCNA 1 RNA and detecting the presence of engineered KCNA 1 RNA in the cell using a hybridisation assay. This method can be carried out in vitro or ex vivo, for example in cell culture or in cells explanted from a human or animal body. Alternatively, the method can be carried out in vivo, for example where the viral particles are administered to a cell in a human or animal subject before extracting the cells or tissues from the human or animal subject in order to detect the presence of engineered KCNA 1 RNA in the cell using a hybridisation assay.

In some embodiments, cells or tissues are extracted from a subject who has been treated with viral particles of the invention in order to examine localisation of the expressed

KCNA 1 gene. Such tissue could be obtained, for instance, from epilepsy surgery in the event of epilepsy gene therapy failure, or post-mortem.

The invention also provides an in vitro or ex vivo method of confirming the presence of engineered KCNA 1 in a cell that has been obtained from a subject administered with a viral particle described herein, the method comprising detecting the presence of engineered KCNA 1 RNA in the cell using a hybridisation assay.

Hybridisation assays are known in the art and generally involve using complementary nucleic acid probes (such as in situ hybridization using labelled probe, Northern blot and related techniques). In some embodiments, the hybridisation assay is an in situ hybridisation assay using a labelled probe, such as a fluorescently labelled probe.

As used herein, the term "probe" refers to a nucleic acid used to detect a complementary nucleic acid. Typically the probe is an RNA probe.

Suitable selective hybridisation conditions for oligonucleotides of 17 to 30 bases include hybridization overnight at 42°C in 6X SSC and washing in 6X SSC at a series of increasing temperatures from 42oC to 65oC. One common formula for calculating the stringency conditions required to achieve hybridization between nucleic acid molecules of a specified sequence homology is (Sambrook et al., 1989): Tm = 81 .5oC + 16.6Log [Na+] + 0.41 (% G+C) - 0.63 (% formamide) - 600/#bp in duplex.

Cells

The invention also provides a cell comprising the nucleic acid or vector described above. In some embodiments, this cell is a mammalian cell such as a human cell. In some embodiments, the cell is a human embryonic kidney cell (HEK) 293. Nucleic acids and sequence variants

The invention also provides a nucleic acid comprising an engineered KCNA 1 gene encoding an edited Kv1 .1 potassium channel, as defined herein. The engineered KCNA 1 gene present in the nucleic acid and edited Kv1 .1 potassium channel can have the requisite features and sequence identity as described herein in relation to the expression vectors.

Alignment and calculation of percentage amino acid or nucleotide sequence identity can be achieved in various ways know to a person of skill in the art, for example, using publically available computer software such as ClustalW 1 .82, T-coffee or Megalign

(DNASTAR) software. When using such software, When using such software, the default parameters, e.g. for gap penalty and extension penalty, are preferably used. The default parameters of ClustalW 1 .82 are: Protein Gap Open Penalty = 10.0, Protein Gap

Extension Penalty = 0.2, Protein matrix = Gonnet, Protein/DNA ENDGAP = -1 ,

Protein/DNA GAPDIST = 4.

The percentage identity can then be calculated from the multiple alignment as (N/T) * 100, where N is the number of positions at which the two sequences share an identical residue, and T is the total number of positions compared. Alternatively, percentage identity can be calculated as (N/S) * 100 where S is the length of the shorter sequence being compared. The amino acid/polypeptide/nucleic acid sequences may be synthesised de novo, or may be native amino acid/polypeptide/nucleic acid sequence, or a derivative thereof. Due to the degeneracy of the genetic code, it is clear that any nucleic acid sequence could be varied or changed without substantially affecting the sequence of the protein encoded thereby, to provide a functional variant thereof. Suitable nucleotide variants are those having a sequence altered by the substitution of different codons that encode the same amino acid within the sequence, thus producing a silent change. Other suitable variants are those having homologous nucleotide sequences but comprising all, or portions of, sequence which are altered by the substitution of different codons that encode an amino acid with a side chain of similar biophysical properties to the amino acid it substitutes, to produce a conservative change. For example small non-polar, hydrophobic amino acids include glycine, alanine, leucine, isoleucine, valine, proline, and methionine. Large non-polar, hydrophobic amino acids include phenylalanine, tryptophan and tyrosine. The polar neutral amino acids include serine, threonine, cysteine, asparagine and glutamine. The positively charged (basic) amino acids include lysine, arginine and histidine. The negatively charged (acidic) amino acids include aspartic acid and glutamic acid. As described herein, suitable variants of the edited Kv1 .1 potassium channel can contain amino acid substitutions at any amino acid other than the amino acid residue at a position corresponding to amino acid residue 400 shown in SEQ ID NO: 2.

Any sub-titles herein are included for convenience only, and are not to be construed as limiting the disclosure in any way. The invention will now be further described with reference to the following non-limiting

Figures and Examples. Other embodiments of the invention will occur to those skilled in the art in the light of these.

The disclosure of all references cited herein, inasmuch as it may be used by those skilled in the art to carry out the invention, is hereby specifically incorporated herein by cross- reference.

Figures Figure 1 : Characterisation of the visual cortex TeNT model of focal neocortical epilepsy.

A. Representative seizure from an adult male Sprague Dawley rat showing its long duration and evolution over time (see Fig. 2 for representative seizures from Lister Hooded and Long-Evans rats). Expanded sections are taken at the times indicated. B. Number of seizures per week for 9 animals recorded over a period of 5 weeks showing an increase to plateau at week 3, followed by a decline in frequency during week 5.

C. Cumulative seizure frequency (per day) over the same period.

D. Seizure duration (per week) over the same period indicating an initial rise to a stable duration of around 100 s.

E. Behavioural correlates of 102 randomly selected electrographic seizures (N = 8 animals, n = 102 seizures). All behaviour was assessed by a neurologist using video recordings time-locked to the ECoG trace. In the 1 1 cases where the behavioural correlate is 'unknown', either the animals were not visible beneath bedding or the video signal was interrupted.

Data are presented as mean ± the standard error of the mean (SEM).

Figure 2: The TeNT visual cortex model of focal neocortical epilepsy is not strain- specific.

Representative seizures from Lister Hooded, Long-Evans and Sprague Dawley rats. Figure 3: Kv1.1 overexpression reduces seizure frequency in the visual cortex model of focal neocortical epilepsy.

A. Neuronal transduction by the CMV-KCNA 1 lentivector is restricted to a narrow column of cortex surrounding the site of injection.

B. Representative seizures from two animals after treatment with the CMV-KCNA 1 lentivector (bottom trace) or its GFP-only control (top trace).

C. Raster plots of all seizures experienced by the same animals over the full 5 weeks of recording. Seizures presented in panel B are marked by asterisks.

D. Seizure frequency (per week) for rats treated with the CMV-KCNA 1 lentivector (n = 8) or its GFP-only control (n = 8). The numbers of seizures experienced each week were normalized to the number experienced by each animal in the week preceding treatment (week Bl). The CMV-KCNA 1 lentivector significantly reduced normalized seizure frequency in the weeks following treatment.

E. Cumulative seizure frequency (per day) for rats treated with the CMV-KCNA 1 lentivector or its GFP-only control. Cumulative seizure counts were normalized to the number experienced by each animal in the 7 days preceding treatment.

Data are presented as mean ± SEM. Figure 4: Kv1.1 overexpression with the CMV-A CMA7 lentivector suppresses inter- ictal spiking

A. Example ECoG traces of inter-ictal spikes.

B. Cumulative inter-ictal spike frequency (per day) for rats treated with the CMV-KCNA 1 lentivector (n=8) or its GFP-only control (n=7). Cumulative inter-ictal spike counts were normalized to the number experienced by each animal in the 7 days preceding treatment. The frequency of inter-ictal spikes was significantly reduced by treatment with the CMV- KCNA 1 lentivector (cumulative values at day 28 compared with a Mann Whitney U test; p = 0.04). Figure 5: Design and characterisation of an optimized KCNA1 gene therapy for clinical translation.

A. Lentiviral transfer plasmid maps for the CMV-KCNA 1 pilot vector (i), the optimized EKC vector (ii) and its reporter-only control (iii). Abbreviations: RSV - Rous sarcoma virus promoter; LTR - long terminal repeat; HIV-1 Ψ - HIV-1 packaging signal; RRE - Rev response element; cPPT/CTS - central polypurine tract and central termination sequence; EF1 a - elongation factor 1 a promoter; WPRE - woodchuck hepatitis virus post-transcriptional regulatory element.

B. Heterologous expression of functional Kv1.1 channels from the optimized EKC transfer plasmid. (i): Representative current-time trace from a Neuro-2a cell transfected with the EKC transfer plasmid. (ii): Plot of mean current density against voltage for cells transfected with the EKC transfer plasmid (Kv; n=13), cells transfected with the dscGFP- only control plasmid (G; n=8), and untransfected controls (UT; n=10). Inset: histogram showing differences in current density between the three groups during the voltage step to +20 mV (Kv vs. UT: p=0.0013; Kv vs. G: p=0.0012; UT vs. G: p=0.82; ns = not significant; Welch's one-way ANOVA with Games-Howell post-hoc tests), (iii): Plot of mean normalised conductance against voltage for cells transfected with the EKC transfer plasmid. Data are fit with a single Boltzmann function. The V0.5 (voltage of half-maximal conductance) of -28.2 mV is similar to values obtained from human embryonic kidney 293 (HEK293) cells transfected with CMV-driven, wild-type KCNA 1 (-32.8 ± 0.9 mV)(Tomlinson et al., 2013). All error bars represent SEM.

C. Bright-field (left) and fluorescence (right) images of a brain slice from a rat injected in the left visual cortex with 1.25 μΙ (~3 x 10 6 infectious units (III)) of the EKC lentivector showing a pattern of transduction similar to that of the CMV-KCNA 1 vector.

D. Immunohistochemical assessment of the cell type specificity of EKC expression, (i): There was no overlap between lentivector-transduced neurons expressing dscGFP and astrocytes stained for GFAP. (ii): There was 100% overlap between dscGFP+ cells and neurons stained for NeuN. (iii): Minimal overlap was observed between dscGFP+ cells and inhibitory interneurons stained for GAD67.

Figure 6: Cortical spread of EKC lentivector transduction.

Bright-field and fluorescence images of 6 sequential left-hemisphere visual cortex slices (70 μηη thick) from a rat brain injected with 1.25 μΙ (~3 x 10 6 IU) of the EKC lentivector. Slices are ordered from top left (rostral) to bottom right (caudal). Scale bars represent 600 μηη and 200 μηη for bright-field and fluorescence images, respectively. Figure 7: EKC gene therapy robustly reduces seizure frequency in a blinded, randomised, pre-clinical trial.

A. Representative seizures from two animals after treatment with the EKC lentivector (bottom trace) or its dscGFP-only control (top trace).

B. Raster plots of all seizures experienced by the same animals over the full 5 weeks of recording. Seizures presented in panel A are marked by asterisks.

C. Normalized seizure frequency (per week) for rats treated with the EKC lentivector (n = 7) or its reporter-only control (n = 1 1 ). The EKC lentivector significantly reduced seizure frequency in the weeks following treatment.

D. Normalized cumulative seizure frequency (per day) for rats treated with the EKC lentivector or its reporter-only control.

Data are presented as mean ± SEM.

Examples Example 1 - Demonstration of delivery of an engineered potassium channel gene in epilepsy gene therapy

Materials and Methods Molecular biology

Lentiviral transfer plasmids were constructed using standard subcloning techniques. KCNA 1 was codon optimized for human expression using GeneOptimizer® software, and synthesised using GeneArt® (Thermo Fisher Scientific). All plasmids were fully sequenced before use.

Voltage clamp recordings Neuro-2a cells were obtained as a gift from the laboratory of S. Hart (UCL Institute of Child Health). Cells were grown in Gibco® Dulbecco's Modified Eagle Medium (DMEM) + GlutaMAX™ (Thermo Fisher Scientific), supplemented with 10% heat-inactivated foetal bovine serum (Thermo Fisher Scientific), 1 % penicillin/streptomycin (Thermo Fisher Scientific) and 1 % non-essential amino acids (Sigma). Cultures were maintained in logarithmic growth phase in a humidified 5% CO2 atmosphere at 37°C. Transfections were performed using TurboFect™ transfection reagent (Thermo Fisher Scientific) according to the manufacturer's instructions. Transfected cells were plated onto 13 mm borosilicate glass coverslips (VWR). Coverslips were placed into the chamber of a BX51 WI fixed-stage upright microscope equipped with UMPLFLN 10x and LUMPLFLN

40x water-immersion objectives (Olympus). Coverslips were submerged in a static bath of extracellular solution with the following composition (in mM): 140 NaCI, 4 KCI, 1 .8 CaC , 2 MgCI 2 , 10 HEPES (pH 7.35, osmolarity -301 mOsm/L). Filamented borosilicate glass micropipettes (GC150-F; Warner Instruments) were pulled to tip resistances between 2.0 and 3.0 ΜΩ using a P-97 Flaming/Brown micropipette puller (Sutter Instrument

Company). Micropipettes were filled with an intracellular solution of the following composition (in mM): 140 KCI, 10 HEPES, 10 EGTA (pH 7.35, osmolarity -291 mOsm/L). Transfected cells were identified by their expression of the fluorescent marker dscGFP, excited with light of wavelength 488 nm. Macroscopic currents were recorded under voltage clamp using the whole-cell patch clamp configuration. The voltage step protocol used was as follows: cells were held at a resting potential of -80 mV and currents evoked by 200 ms depolarising steps delivered in 10 mV increments up to +20 mV. A 40 ms hyperpolarising step to -100 mV was included before returning to baseline. Data were filtered at 3 kHz and acquired at 10 kHz using WinWCP software (J. Dempster, University of Strathclyde) and an Axon Multiclamp 700B amplifier (Molecular Devices). Series resistance compensation was employed throughout, with prediction and correction components adjusted to 80% and the bandwidth set to 1.2 kHz. Cells with series resistance greater than 10 ΜΩ were excluded from the analysis. All recordings were made at room temperature (23-26°C). The liquid junction potential, calculated to be +4.1 mV, was left uncorrected. Leak currents were minimal and left unsubtracted.

For analysis, evoked currents were taken as the steady-state current in the last 40 ms of each voltage step. Baseline holding currents were subtracted before division by cell capacitance to generate current density values. To calculate normalised conductance, the current density at each voltage step was divided by the step potential minus the potassium reversal potential (-91.34 mV). This generates raw conductance values that are corrected for the variation in K + driving force which accompanies stepwise changes in membrane potential. Plots of raw conductance against voltage for each EKC-transfected cell were fit with individual Boltzmann functions given by the equation:

1 + e k

where G is the conductance, V the voltage, Ai the initial (minimum) conductance, A2 the final (maximum) conductance, V0.5 the voltage of half-maximal conductance, and k the slope factor. Raw conductance values were normalised to Ai and A 2 of their own

Boltzmann functions. Raw conductance values were normalised to Ai and A2 of their own Boltzmann functions. Normalised conductance was then plotted against voltage for each EKC-transfected cell and again fit with individual Boltzmann functions. The mean adjusted R 2 and V0.5 values presented in the results were extracted from these fits. For Fig. 5Biii, mean normalised conductance across cells was plotted against voltage before fitting with a final Boltzmann function. In line with convention, adjusted R 2 and V0.5 values for this fit were not reported.

Lentiviral synthesis

The CNW/-KCNA 1 lentivector was identical to that used in Wykes et al., 2012 and has the nucleotide sequence shown in SEQ I D NO: 6.

HEK293 producer cells were grown in Gibco® DMEM + GlutaMAX™, supplemented with 10% heat-inactivated foetal bovine serum and 1 % penicillin/streptomycin. Cultures were maintained in logarithmic growth phase in a humidified 5% CO2 atmosphere at 37°C.

Cells were split every 3-4 days using 0.05% Trypsin-EDTA (Thermo Fisher Scientific) and never grown for more than 15 passages. For lentiviral synthesis, cells were grown to a confluency of -70% and transiently co-transfected with pMDG-VSV.G, pCMVdR8.74 D64V , and either the EKC transfer plasmid or its dscGFP-only control. pMDG-VSV.G and pCMVdR8.74 D64V were obtained as gifts from the laboratory of A.J. Thrasher and W.

Qasim (UCL Institute of Child Health). pMDG(VSV-G) encodes the envelope glycoprotein of the vesicular stomatitis virus. Lentivectors pseudotyped with VSV-G display a broad tropism and are relatively resistant to mechanical destruction during ultracentrifugation (Burns et al., 1993). pCMVdR8.74 D64V is a 2 nd generation human immunodeficiency virus 1 (H IV-1 ) packaging plasmid with an aspartic acid 64 -valine mutation in its integrase coding sequence. This mutation reduces genomic integration by a factor of 10,000 compared to wild-type levels (Leavitt et al., 1996). The mass ratio of envelope to packaging to transfer plasmids was 1 : 2.5: 1 .5. Transfections were performed using Lipofectamine® 2000 transfection reagent (Thermo Fisher Scientific) at approximately 160 μΙ per 100 μg of total plasmid DNA. The transfection medium was removed after 18 hours and replaced with fresh medium. Two media harvests were collected, at 40 hours and 60 hours following transfection. Special care was taken to minimise cell detachment during each harvest. Harvested media were pre-cleaned by centrifugation at 1000 rpm for 3 min at 4 °C, filtered through 0.45 μηη micropores and stored at 4 °C. Media were overlaid on a sucrose solution with the following composition (in mM): 50 Tris-HCI, 100 NaCI, 0.5 EDTA (pH 7.4, 10% w/v sucrose), and centrifuged at 20,000 rpm for 2 hr at 4 °C. The supernatant was discarded and lentiviral pellet resuspended in sterile PBS. Lentiviral suspensions were aliquoted and snap-frozen in liquid nitrogen before long-term storage at -80 °C. A rough approximation of viral titre was obtained using the Lenti-X™ p24 rapid titer kit (Clontech). Each titration was performed in triplicate with 3 separate aliquots of each virus. Estimated titres were 2.42 χ 10 9 l U/ml (EKC) and 4.26 * 10 9 l U/ml (control).

Surgical procedures

All experiments were performed in accordance with the United Kingdom Animal (Scientific Procedures) Act 1986. Adult male rats (Sprague Dawley, Long-Evans or Lister Hooded;

300-400g) were anaesthetized and placed into a stereotaxic frame (Kopf). 15 ng of TeNT (a gift from the laboratory of G. Schiavo (UCL Institute of Neurology)) was injected into layer 5 of the right visual cortex in a final volume of 1.0 μΙ at a rate of 100 nl rnin -1 (coordinates: 3 mm lateral, 7 mm posterior of bregma; 1 .0 mm deep from the pia). An ECoG transmitter (A3028E; Open Source Instruments, MA, USA) was implanted subcutaneously with a subdural intracranial recording electrode positioned above the injection site. A reference electrode was implanted in the contralateral skull. A cannula (Plastics One) was positioned above the injection site for delivery of lentiviral vectors 1 1 or 14 days later. Each rat received a maximum of 2.0 μΙ of lentivirus injected directly into the seizure focus. Animals injected with TeNT were housed separately in Faraday cages for the duration of the study.

ECoG acquisition and analysis

ECoG was recorded continuously for up to 6 weeks after surgery. Data were acquired using hardware and software purchased from Open Source Instruments. The method of seizure detection differed for the characterisation of the model, the pilot study and the final preclinical experiment. For the model characterisation, seizures were detected by continuous observation of the entire ECoG dataset by a neurologist. For the pilot study, ECoG traces were first divided into 1 s epochs. Four metrics (power, coastline, intermittency and coherence) were quantified for each epoch, and their values compared to those from a user-curated library of epochs validated by video as representing seizure activity (Wykes et al., 2012). Matched values were fed into a consolidation script that returned all instances of 5 or more sequential epochs identified as containing seizure activity. All seizures in the consolidation output were verified by an experimenter. For the final preclinical trial, 6 metrics were quantified for each epoch (power, coastline, intermittency, coherence, asymmetry and rhythm) and all matched values were checked for seizure activity without the use of a consolidation script. Seizure counts in this experiment were performed by an experimenter blinded to the treatment. For all datasets the minimum duration for a seizure was set at 10 s. Behavioural seizure assessment

Assessment of the behavioural correlates of electrographic seizures was carried out by a neurologist. Seizures were randomly selected from the 5 weeks of recording, and associated behaviours observed using top-down video footage time-locked to the ECoG trace.

Immunohistochemistry

1 week after lentivirus injection rats were terminally anaesthetised with sodium pentobarbital (Euthatal; Merial) and transcardially perfused with cold (4 °C) heparinised PBS (80 mg/litre heparin sodium salt; Sigma) followed by 4% paraformaldehyde (PFA) in PBS (Santa Cruz Biotechnology). Brains were removed and post-fixed in 4% PFA at 4 °C for a further 24 hours. After washing in PBS brains were sliced into 70 μηη coronal sections using a vibrating microtome (Leica). Slices were stored free-floating at 4 °C in PBS plus 0.02% sodium azide (Sigma). For antibody staining, slices were permeabilised for 20 minutes in PBS plus 0.3% Triton X-100 (Sigma) before blocking for 1 hour in PBS plus 0.3% Triton X-100 (Sigma), 1 % bovine serum albumin (Sigma) and 4% goat serum (Sigma). Slices were incubated overnight at 4 °C in PBS plus 0.3% Triton X-100 and rabbit anti-NeuN (diluted 1 :750; ab177487; Abeam), mouse anti-GFAP (diluted 1 :500; MAB3402; Merck Millipore) or mouse anti-GAD67 (diluted 1 :500; MAB5406; Merck Millipore) primary antibodies. After three 10 minute washes in PBS, slices were incubated at room temperature for 3 hours in PBS plus the relevant Alexa Fluor® 594-conjugated secondary antibody (goat anti-rabbit (A-1 1037; Thermo Fisher Scientific) or goat anti- mouse (A-1 1005; Thermo Fisher Scientific); both diluted 1 :750). After a further three 10 minute washes in PBS, slices were mounted onto plain glass microscope slides (Thermo Fisher Scientific) using Vectashield® HardSet™ mounting medium (Vector Laboratories) and borosilicate glass coverslips (VWR). Bright-field and fluorescence images were acquired using one of two microscopes: an Axio Imager A1 fluorescence microscope (Axiovision LE software) equipped with 2.5x, 10x and 40x EC Plan-Neofluar non- immersion objectives, or an inverted LSM 710 confocal laser scanning microscope (ZEN 2009 software) equipped with 40x and 63x EC Plan-Neofluar oil-immersion objectives (Zeiss). For the confocal microscope, dscGFP and Alexa Fluor® 594 were excited with the 488 nm and 561 nm lines of an argon or diode pumped solid state (DPSS) laser, respectively. All image processing was performed using ImageJ software. Composite images were assembled using the MosaicJ ImageJ plugin.

Statistics

Efficacy of treatment data (Fig. 3D, 7C) were analysed using a generalized log-linear mixed model with random effect of animal (autoregressive covariance) and fixed effects of treatment group, week, and the interaction between treatment group and week.

Seizure counts in the week preceding treatment (Fig. 7C) were compared using a Mann Whitney U test. Current densities at +20 mV (Fig. 5Bii) were compared using a Welch's one-way ANOVA followed by Games-Howell post-hoc tests.

Results

Injection of TeNT into the visual cortex produces a model of focal epilepsy with discrete seizures

Starting with a model developed in mice (Mainardi et al., 2012), we characterized the electrographic features, temporal evolution, and behavioural correlates of seizures evoked by injection of TeNT into the visual cortices of 9 adult male Sprague Dawley rats. In striking contrast to the brief, near-continuous epileptiform discharges seen following TeNT injection into the motor cortex (Wykes et al., 2012), injection into the visual cortex produced discrete spontaneous seizures (Fig. 1A). Seizures emerged approximately 4 days after the injection of TeNT, increased in frequency over 7-10 days, reached a plateau for 2-3 weeks, and resolved in most animals after 5 or 6 weeks (Fig. 1 B, C). The total number of seizures experienced by each animal was highly variable. Average seizure duration evolved over time, increasing from just under 50 s in the first week to approximately 100 s for the next four (Fig. 1 D). Time-locked video-ECoG was employed to identify the behavioural correlates of a randomly selected subset of seizures (n = 102; Fig. 1 E). For 1 1 seizures (10.8%), associated behaviours were unobservable due to interrupted recordings or the animal remaining out of sight within its environmental enrichment material. All remaining seizures had observable behavioural correlates. For 45 seizures (44.1 %) these were subtle, including repetitive eye blinking, sudden increases in agitation or aggressive searching behaviour. Unilateral motor involvement, e.g. twitching of the contralateral limb, was observed for 19 seizures (18.6%), with bilateral motor symptoms seen in a further 6 (5.9%). 21 seizures (20.6%) developed into full bilateral tonic-clonic seizures. This model of focal neocortical epilepsy was not strain- specific; similar seizures were evoked in both Lister Hooded and Long-Evans rats (Fig. 2).

A pilot study shows KCNA1 gene therapy is sufficient to reduce spontaneous seizures

We asked whether the CMV-driven Kv1.1 vector (CMV-KCNA 1) used earlier in a model of EPC (Wykes et al., 2012) remained effective against these longer and more complex seizures. Two weeks after the injection of TeNT, following the establishment of spontaneous seizures, animals were randomly divided into 2 groups and injected via pre- implanted cannula with either the CMV-KCNA 1 lentivector or a control lentivector expressing only green fluorescent protein (GFP). ECoG recordings were continued for a further 4 weeks. As previously observed (Wykes et al., 2012), the CWW/-KCNA 1 lentivector transduced neurons within a narrow column of the cortex (Fig. 3A). Given the high variability in total seizure counts, to compare seizure frequency between the two treatment groups the numbers of seizures experienced each week were normalized to the number experienced by each animal in the week preceding treatment (baseline week (Bl)). The CMV-KCNA 1 lentivector significantly reduced normalized seizure frequency in the weeks following treatment (generalized log-linear mixed model on weeks 0-3, treatment * week interaction effect: F(1 ,60) = 69.499, p < 0.001 ; Fig. 3D). The therapeutic effect emerged rapidly; plots of cumulative daily seizure frequency for the two treatment groups begin to diverge within 3 days of lentivector injection (Fig. 3E). Kv1.1

overexpression did not influence seizure duration (data not shown).

Epilepsy is associated with a number of comorbidities. Among these are cognitive deficits which in temporal lobe epilepsies strongly correlate to inter-ictal discharges (Holmes GL 2013, Bragatti JA et al 2014, Dinkelacker V et al 2016). Similarly, in the neocortex inter- ictal activity is likely to cause significant disruption to normal brain function. In addition to its suppression of seizure frequency, Kv1.1 overexpression with the CMV-KCNA 1 lentivector significantly reduced the frequency of inter-ictal discharges, another signature consistent with epileptogenesis (Fig. 4).

Because our model of focal neocortical epilepsy was not strain-specific (Fig. 2), this initial study included 16 animals from 3 different strains (Sprague Dawley, Lister Hooded and Long-Evans). To ensure that the positive effect of CMV-KCNA 1 treatment was not biased by the minority of Lister Hooded and Long-Evans animals, seizure frequency data were reanalysed for the Sprague Dawley animals only. Despite the small sample size (6 treated vs. 5 controls), the CMV-KCNA 1 lentivector still significantly reduced normalized seizure frequency in the weeks following treatment (generalized log-linear mixed model on weeks 0-3, treatment * week interaction effect: F(1 ,40) = 4.851 , p = 0.033). This pilot study therefore strongly suggests that over-expression of KCNA 1 is sufficient to reduce the number of discrete spontaneous seizures. Design and characterisation of a gene therapy tool optimized for clinical translation

To bring KCNA 1 gene therapy closer to the clinic, we designed a new lentiviral transfer plasmid (Fig. 5Aii). The CMV promoter from the original CMV-KCNA 1 construct (Fig. 5Ai) was replaced with a 1 .3 kb human CAMK2A promoter (hCAMK2A) to bias expression to excitatory neurons (Dittgen et al., 2004; Yaguchi et al., 2013). The KCNA 1 gene was codon-optimized for expression in human cells, and mutated to introduce an I400V amino acid change normally generated by RNA-editing. The I400V substitution elicits a 20-fold increase in the rate at which Kv1 .1 channels recover from inactivation (Bhalla et al., 2004). For preclinical evaluation, a dscGFP reporter gene was linked to the engineered potassium channel (EKC) gene by a T2A peptide, which permits dual protein expression from a single promoter.

The salient components of our EKC lentiviral transfer plasmid (Fig. 5Aii) include:

1 ) A 1 .3kb human CAMK2A promoter (hCAMK2A) comprising 100 bp of the 5'

untranslated region of CAMK2A coupled to 1.2 kb of upstream DNA. A murine Camk2a promoter of similar size has been shown to drive pyramidal neuron- specific gene expression in lentiviral-injected rat barrel cortex (Dittgen et al., 2004) and primate motor cortex (Yaguchi et al., 2013). This contrasts with the CMV promoter of our pilot vector, which is known to drive transgene expression preferentially within glia following lentiviral injection into the primate rhinal (Lerchner et al., 2014) or motor (Yaguchi et al., 2013) cortices.

2) An engineered potassium channel gene consisting of a codon-optimized KCNA 1 with an adenine 1998 >guanine point mutation to modify channel inactivation properties. Codon optimization is useful beyond the benefit of increased Kv1.1 expression. It permits specific tracking of transgene expression with RNA-targeted techniques that would otherwise fail to distinguish between transgenic and endogenous KCNA 1. Such tracking will prove particularly useful further along the translation pipeline with vectors that necessarily lack coding sequences for fluorescent reporter proteins. At negative membrane potentials, channels containing unedited I 400 recover from inactivation at a rate around twenty times slower than their edited (V 400 ) counterparts (Bhalla et al., 2004). Because Kv1 .1 channels that recover faster from inactivation would be expected to dampen neuronal excitability, we decided to pre-empt the RNA edit in our codon-optimized KCNA 1 by purposefully introducing the A 1998 G point mutation.

3) A superbright dscGFP reporter gene linked to EKC by a T2A peptide. 2A peptides are short (-20 amino acids) sequences that permit multicistronic gene expression from single promoters by impairing peptide bond formation during ribosome- mediated translation (Szymczak and Vignali, 2005). Importantly, genes either side of a 2A peptide are expressed in a 1 :1 ratio with translation taking place simultaneously. As such, reporter expression serves as a very reliable indicator of therapeutic transgene expression. This set-up contrasts with that of our pilot vector, where therapeutic and reporter transgenes were placed under the transcriptional control of separate and distinct promoters. The EKC transfer plasmid used in these experiments has the sequence shown in SEQ ID NO: 7. The sequence of the dscGFP-only control transfer plasmid has the sequence shown in SEQ ID NO: 8. To determine whether the optimized transfer plasmid could be decoded to produce functional Kv1 .1 channels, Neuro-2a cells were transiently transfected with the EKC or dscGFP-only control transfer plasmids and subjected to whole-cell patch clamping. Cells exposed to the transfection mixture but lacking dscGFP fluorescence were used as a second set of controls. A typical voltage step protocol evoked negligible outward currents in both control groups, while Neuro-2a cells transfected with the EKC transfer plasmid displayed large voltage-dependent currents that peaked in amplitude at several nanoamps (Fig. 5Bi,ii). Current densities at the largest depolarising voltage step (+20 mV) were significantly larger in EKC-transfected cells than controls, which displayed only small endogenous voltage-dependent currents (Fig. 5Bii). There was no significant difference in current density between untransfected cells and cells transfected with the control plasmid (Fig. 5Bii). To determine if our molecular optimizations influenced the voltage dependence of Kv1.1 channel activation, normalised conductance-voltage plots were fit with individual Boltzmann functions for each EKC-transfected cell. A mean adjusted R 2 of 0.995 ± 0.0007 demonstrated a good fit across all cells. The mean V0.5 of -28.2 ± 0.4 mV was similar to values obtained from HEK293 cells transfected with CMV- driven, wild-type KCNA 1 (-32.8 ± 0.9 mV) (Tomlinson et al., 2013). A plot of mean normalised conductance against voltage for all SKC-transfected cells, fit with its own Boltzmann function, is provided in Fig. 5Biii. Together these data indicate that the optimized EKC transfer plasmid supports the generation of robust Kv1 .1 currents.

The EKC transfer plasmid was packaged into a non-integrating lentiviral vector (Yafiez- Mufioz et al., 2006). When injected into the rat visual cortex, this lentivector drove strong, localised expression of the dscGFP reporter (Fig. 5C). Imaging of sequential brain slices produced an estimated transduction volume of -0.075 mm3 (Fig. 6).

Immunohistochemistry revealed no visible overlap between dscGFP expression and glial fibrillary acidic protein (GFAP) staining (0/512 dscGFP+ cells co-localized with GFAP, n = 3 animals; Fig. 5Di). In contrast, all dscGFP+ cells co-localized with the neuronal marker NeuN (714/714, n = 3 animals; Fig. 5Di). These data indicate that transgene expression from the EKC lentivector is restricted to neurons . There was minimal overlap between dscGFP expression and staining for glutamic acid decarboxylase 67 (GAD67), an enzymatic marker for GABAergic neurons (3/603 dscGFP+ cells co-localized with GAD67, n = 3 animals; Fig. 3Diii). This suggests that EKC transgene expression is largely restricted to excitatory neurons. EKC gene therapy reduces seizure frequency in a randomised, blinded, preclinical trial

To test the efficacy of the EKC lentivector, we designed a randomised, blinded preclinical trial to mimic clinical trial conditions, and selected reduced seizure frequency as the primary outcome measure. A new batch of TeNT with slightly reduced potency produced fewer overall seizures and improved animal welfare. Eleven days after the injection of TeNT, 26 Sprague Dawley rats were randomly divided into 2 groups and injected via pre- implanted cannula with either the EKC lentivector or a dscGFP-only control vector. ECoG recordings were continued for a further 4 weeks. The timeline was altered from that of the pilot study to treat after 1 1 days in order to capture the period when seizure activity is at its highest (2-4 weeks following TeNT). To minimise the confounding influence of animals that failed to develop robust epilepsy, subjects were excluded if they experienced fewer than 5 seizures in the week preceding treatment. This criterion, applied prior to unblinding, led to the exclusion of 8 animals (6 EKC, 2 control). There was no significant difference between treatment groups in the number of seizures experienced in the week preceding virus injection (Mann Whitney U test, p = 0.185). Analysis of the primary outcome measure indicated that EKC treatment produced a robust decrease in seizure frequency over time (generalized log-linear mixed model on weeks 0-3, treatment * week interaction effect: F(1 ,67) = 29.704, p < 0.001 ; Fig. 7C). As in the pilot study, the effect lasted for the duration of recording, and the absolute effect size only decreased as seizures abated in the control group. Again, the therapeutic effect emerged rapidly, with plots of cumulative daily seizure frequency for the two groups starting to diverge 2 days after treatment (Fig. 7D).

Discussion

EKC gene therapy represents an effective new treatment for focal neocortical seizures in a format adapted to improve safety and translation to the clinic.

We have previously shown that overexpression of Kv1.1 can reduce the frequency of epileptiform discharges in a motor cortex TeNT model of EPC (Wykes et al., 2012).

However, it was unclear whether Kv1 .1 overexpression would be sufficient to inhibit discrete long-lasting seizures. We show here, in two independent experiments, that Kv1.1 overexpression is indeed sufficient to reduce seizure frequency. The effect on seizure number is pronounced, but once initiated the seizures progress in a similar pattern with similar electrographic features in both treated and control animals.

Injection of TeNT into the occipital cortex induced seizures that lasted markedly longer (50-150s) than those induced by TeNT injection in motor cortex (<1 s) (Wykes et al., 2012). The difference, which parallels that seen with occipital lobe seizures and EPC in human patients, may be a consequence of different connectivity in the occipital and motor cortices. Further studies will be needed to determine how cortical architecture impacts the type of epileptiform activity induced by TeNT insult.

Lentiviral gene therapy approaches are becoming more common in CNS disorders, and have shown good safety and tolerability even in extended trials (Palfi et al., 2014). In the case of epilepsy, an additional safety concern is the possibility of potassium channel overexpression in interneurons, which could aggravate seizure activity by exacerbating rather than attenuating local excitability. To mitigate this risk we have utilised a human CAMK2A promoter that in rats leads to very little expression in GABAergic cells. Our estimate of specificity may even be slightly conservative as a small proportion of excitatory pyramidal neurons in the rodent cortex stain positively for GABA as well as glutamate (Hill et al., 2000; Lavdas et al., 1996), and may therefore express GAD67 despite pyramidal morphology and physiology.

Because the role of potassium channels, including Kv1.1 , in regulating neuronal excitability is conserved across a broad range of neurons, potassium channel overexpression may hold therapeutic promise in the treatment of other diseases characterised by neuronal hyperexcitability. There is currently an unmet clinical need for new treatments for chronic pain, and a variety of gene therapy approaches aimed at reducing the excitability of dorsal root ganglion neurons have already demonstrated preclinical efficacy (Snowball and Schorge, 2015). Other disorders such as Parkinson's disease may be associated with excessive activity in specific groups of neurons (Lobb, 2014), and could be candidates for treatment if suitable cell type specific promoters can be identified.

References

Baum, C, von Kalle, C, Staal, F.J.T., Li, Z., Fehse, B., Schmidt, M., Weerkamp, F., Karlsson, S., Wagemaker, G., and Williams, D.A. (2004). Chance or necessity?

Insertional mutagenesis in gene therapy and its consequences. Mol. Ther. J. Am. Soc. Gene Ther. 9, 5-13.

Bhalla, T., Rosenthal, J.J.C., Holmgren, M., and Reenan, R. (2004). Control of human potassium channel inactivation by editing of a small mRNA hairpin. Nat. Struct. Mol. Biol. 11, 950-956. Biffi, A., Montini, E., Lorioli, L, Cesani, M., Fumagalli, F., Plati, T., Baldoli, C, Martino, S., Calabria, A., Canale, S., et al. (2013). Lentiviral Hematopoietic Stem Cell Gene Therapy Benefits Metachromatic Leukodystrophy. Science 341, 1233158.

Bovolenta, R., Zucchini, S., Paradiso, B., Rodi, D., Merigo, F., Navarro Mora, G., Osculati, F., Berto, E., Marconi, P., Marzola, A., et al. (2010). Hippocampal FGF-2 and BDNF overexpression attenuates epileptogenesis-associated neuroinflammation and reduces spontaneous recurrent seizures. J. Neuroinflammation 7, 81 .

Burns, J.C., Friedmann, T., Driever, W., Burrascano, M., and Yee, J.K. (1993). Vesicular stomatitis virus G glycoprotein pseudotyped retroviral vectors: concentration to very high titer and efficient gene transfer into mammalian and nonmammalian cells. Proc. Natl. Acad. Sci. U. S. A. 90, 8033-8037.

Cartier, N., Hacein-Bey-Abina, S., Bartholomae, C.C., Veres, G., Schmidt, M., Kutschera, I., Vidaud, M., Abel, U., Dal-Cortivo, L., Caccavelli, L., et al. (2009). Hematopoietic stem cell gene therapy with a lentiviral vector in X-linked adrenoleukodystrophy. Science 326, 818-823. D'Adamo MC, Catacuzzeno L, Di Giovanni G, Franciolini F, Pessia M. (2013). K(+) channelepsy: progress in the neurobiology of potassium channels and epilepsy. Front. Cell. Neurosci. 7, 134

Devinsky, O. (201 1 ). Sudden, unexpected death in epilepsy. N. Engl. J. Med. 365, 1801- 181 1 . Dittgen, T., Nimmerjahn, A., Komai, S., Licznerski, P., Waters, J., Margrie, T.W.,

Helmchen, F., Denk, W., Brecht, M., and Osten, P. (2004). Lentivirus-based genetic manipulations of cortical neurons and their optical and electrophysiological monitoring in vivo. Proc. Natl. Acad. Sci. U. S. A. 101, 18206-1821 1 .

Galanopoulou, A.S., Buckmaster, P.S., Staley, K.J., Moshe, S.L., Perucca, E., Engel, J., Jr, Loscher, W., Noebels, J.L., Pitkanen, A., Stables, J., et al. (2012). Identification of new epilepsy treatments: issues in preclinical methodology. Epilepsia 53, 571-582. Haberman, R.P., Samulski, R.J., and McCown, T.J. (2003). Attenuation of seizures and neuronal death by adeno-associated virus vector galanin expression and secretion. Nat Med 9, 1076-1080.

Hacein-Bey-Abina, S., Von Kalle, C, Schmidt, M., McCormack, M.P., Wulffraat, N., Leboulch, P., Lim, A., Osborne, C.S., Pawliuk, R., Morillon, E., et al. (2003). LM02- associated clonal T cell proliferation in two patients after gene therapy for SCID-X1 .

Science 302, 415-419.

Heeroma, J.H., Henneberger, C, Rajakulendran, S., Hanna, M.G., Schorge, S., and Kullmann, D.M. (2009). Episodic ataxia type 1 mutations differentially affect neuronal excitability and transmitter release. Dis. Model. Mech. 2, 612-619.

Hill, E., Kalloniatis, M., and Tan, S.S. (2000). Glutamate, GABA and precursor amino acids in adult mouse neocortex: cellular diversity revealed by quantitative

immunocytochemistry. Cereb. Cortex N. Y. N 1991 10, 1 132-1 142.

Hoppe, C, and Elger, C.E. (201 1 ). Depression in epilepsy: a critical review from a clinical perspective. Nat. Rev. Neurol. 7, 462-472.

Kanter-Schlifke, I., Georgievska, B., Kirik, D., and Kokaia, M. (2007). Seizure suppression by GDNF gene therapy in animal models of epilepsy. Mol. Ther. J. Am. Soc. Gene Ther. 15, 1 106-1 1 13.

Kantor, B., McCown, T., Leone, P., and Gray, S.J. (2014). Chapter Two - Clinical Applications Involving CNS Gene Transfer. In Advances in Genetics, J. CD. and S.F.G. Theodore Friedmann, ed. (Academic Press), pp. 71-124.

Katzel, D., Nicholson, E., Schorge, S., Walker, M.C, and Kullmann, D.M. (2014).

Chemical-genetic attenuation of focal neocortical seizures. Nat. Commun. 5, 3847.

Kullmann, D.M., Schorge, S., Walker, M.C, and Wykes, R.C (2014). Gene therapy in epilepsy-is it time for clinical trials? Nat. Rev. Neurol. 10, 300-304.

Kwan, P., Schachter, S.C, and Brodie, M.J. (201 1 ). Drug-resistant epilepsy. N. Engl. J. Med. 365, 919-926.

Lavdas, A.A., Mione, M.C, and Parnavelas, J.G. (1996). Neuronal clones in the cerebral cortex show morphological and neurotransmitter heterogeneity during development. Cereb. Cortex N. Y. N 1991 6, 490-497.

Leavitt, A.D., Robles, G., Alesandro, N., and Varmus, H.E. (1996). Human

immunodeficiency virus type 1 integrase mutants retain in vitro integrase activity yet fail to integrate viral DNA efficiently during infection. J. Virol. 70, 721-728.

Lerchner, W., Corgiat, B., Der Minassian, V., Saunders, R.C, and Richmond, B.J. (2014). Injection parameters and virus dependent choice of promoters to improve neuron targeting in the nonhuman primate brain. Gene Ther. Lin, E.-J.D., Young, D., Baer, K., Herzog, H., and During, M.J. (2006). Differential actions of NPY on seizure modulation via Y1 and Y2 receptors: evidence from receptor knockout mice. Epilepsia 47, 773-780.

Lobb, C. (2014). Abnormal Bursting as a Pathophysiological Mechanism in Parkinson's Disease. Basal Ganglia 3, 187-195.

Lundberg, C, Bjorklund, T., Carlsson, T., Jakobsson, J., Hantraye, P., Deglon, N., and Kirik, D. (2008). Applications of lentiviral vectors for biology and gene therapy of neurological disorders. Curr. Gene Ther. 8, 461-473.

Mainardi, M., Pietrasanta, M., Vannini, E., Rossetto, O., and Caleo, M. (2012). Tetanus neurotoxin-induced epilepsy in mouse visual cortex. Epilepsia 53, e132-136.

McCown, T.J. (2006). Adeno-associated Virus-Mediated Expression and Constitutive Secretion of Galanin Suppresses Limbic Seizure Activity in Vivo. Mol Ther 14, 63-68.

Ngugi, A.K., Bottomley, C, Kleinschmidt, I., Sander, J.W., and Newton, C.R. (2010). Estimation of the burden of active and life-time epilepsy: a meta-analytic approach.

Epilepsia 51, 883-890.

Nikitidou, L., Torp, M., Fjord-Larsen, L., Kusk, P., Wahlberg, L.U., and Kokaia, M. (2014). Encapsulated galanin-producing cells attenuate focal epileptic seizures in the

hippocampus. Epilepsia 55, 167-174.

Noe', F.M., S0rensen, AT., Kokaia, M., and Vezzani, A. (2012). Gene therapy of focal onset epilepsy using adeno-associated virus vector-mediated overexpression of neuropeptide Y. In Jasper's Basic Mechanisms of the Epilepsies, J.L. Noebels, M. Avoli, M.A. Rogawski, R.W. Olsen, and A.V. Delgado-Escueta, eds. (Bethesda (MD): National Center for Biotechnology Information (US)), p.

Palfi, S., Gurruchaga, J.M., Ralph, G.S., Lepetit, H., Lavisse, S., Buttery, P.C., Watts, C, Miskin, J., Kelleher, M., Deeley, S., et al. (2014). Long-term safety and tolerability of ProSavin, a lentiviral vector-based gene therapy for Parkinson's disease: a dose escalation, open-label, phase 1/2 trial. Lancet Lond. Engl. 383, 1 138-1 146.

Picot, M.-C, Baldy-Moulinier, M., Daures, J.-P., Dujols, P., and Crespel, A. (2008). The prevalence of epilepsy and pharmacoresistant epilepsy in adults: a population-based study in a Western European country. Epilepsia 49, 1230-1238.

Rahim, A.A., Wong, A.M.S., Howe, S.J., Buckley, S.M.K., Acosta-Saltos, A.D., Elston, K.E., Ward, N.J., Philpott, N.J., Cooper, J.D., Anderson, P.N., et al. (2009). Efficient gene delivery to the adult and fetal CNS using pseudotyped non-integrating lentiviral vectors. Gene Ther. 16, 509-520. Richichi, C, Lin, E.-J.D., Stefanin, D., Colella, D., Ravizza, T., Grignaschi, G.,

Veglianese, P., Sperk, G., During, M.J., and Vezzani, A. (2004). Anticonvulsant and antiepileptogenic effects mediated by adeno-associated virus vector neuropeptide Y expression in the rat hippocampus. J. Neurosci. Off. J. Soc. Neurosci. 24, 3051-3059.

Schuele, S.U., and Luders, H.O. (2008). Intractable epilepsy: management and therapeutic alternatives. Lancet Neurol. 7, 514-524. Snowball, A., and Schorge, S. (2015). Changing channels in pain and epilepsy: Exploiting ion channel gene therapy for disorders of neuronal hyperexcitability. FEBS Lett. 589, 1620-1634.

Streit AK, Derst C, Wegner S, Heinemann U, Zahn RK, Decher N. (201 1 ) RNA editing of Kv1 .1 channels may account for reduced ictogenic potential of 4-aminopyridine in chronic epileptic rats. Epilepsia. 52, 645-8

Streit AK, Matschke LA, Dolga AM, Rinne S, Decher N. (2014). RNA editing in the central cavity as a mechanism to regulate surface expression of the voltage-gated potassium channel. J Biol Chem. 289, 2<6Ί<62-Ί

Szymczak, A.L., and Vignali, D.A.A. (2005). Development of 2A peptide-based strategies in the design of multicistronic vectors. Expert Opin. Biol. Ther. 5, 627-638.

Tomlinson, S.E., Rajakulendran, S., Tan, S.V., Graves, T.D., Bamiou, D.E., Labrum, R.W., Burke, D., Sue, CM., Giunti, P., Schorge, S., et al. (2013). Clinical, genetic, neurophysiological and functional study of new mutations in episodic ataxia type 1 . J Neurol Neurosurg Psychiatry. Woldbye, D.P.D., Angehagen, M., G0tzsche, C.R., Elbrand-Bek, H., S0rensen, AT.,

Christiansen, S.H., Olesen, M.V., Nikitidou, L., Hansen, T.V.O., Kanter-Schlifke, I., et al. (2010). Adeno-associated viral vector-induced overexpression of neuropeptide Y Y2 receptors in the hippocampus suppresses seizures. Brain J. Neurol. 133, 2778-2788.

Wykes, R.C, Heeroma, J.H., Kullmann, D.M., Walker, M.C, and Schorge, S. (201 1 ) Lentiviral-mediated over-expression of the potassium channel Kv1.1 as a treatment for focal neocortical epilepsy. Poster No. 248.20/S6. 201 1 Neuroscience Meeting Planner. Washington, DC: Society for Neuroscience, 201 1 . Online.

Wykes, R.C, Heeroma, J.H., Mantoan, L., Zheng, K., MacDonald, D.C, Deisseroth, K., Hashemi, K.S., Walker, M.C, Schorge, S., and Kullmann, D.M. (2012). Optogenetic and potassium channel gene therapy in a rodent model of focal neocortical epilepsy. Sci. Transl. Med. 4, 161 ra152.

Wykes, R.C. and Lignani. G (2018). Gene therapy and editing: Novel potential treatments for neuronal channelopathies. Neuropharmacology. 132, 108-1 17

Yaguchi, M., Ohashi, Y., Tsubota, T., Sato, A., Koyano, K.W., Wang, N., and Miyashita, Y. (2013). Characterization of the properties of seven promoters in the motor cortex of rats and monkeys after lentiviral vector-mediated gene transfer. Hum. Gene Ther.

Methods 24, 333-344. Yanez-Munoz, R.J., Balaggan, K.S., MacNeil, A., Howe, S.J., Schmidt, M., Smith, A.J., Buch, P., MacLaren, R.E., Anderson, P.N., Barker, S.E., et al. (2006). Effective gene therapy with nonintegrating lentiviral vectors. Nat. Med. 12, 348-353.

Sequence Annex

Nucleotide sequence of an exemplary engineered human KCNA 1 gene (SEQ ID NO: 1 )

ATGACCGTGATGAGCGGCGAGAACGTGGACGAGGCCTCTGCCGCTCCTGGACACCCT CAGGATGGC AGCTATCCCAGACAGGCCGACCACGACGATCACGAGTGCTGCGAGCGGGTCGTGATCAAC ATCAGC GGCCTGAGATTCGAGACACAGCTGAAAACCCTGGCCCAGTTCCCCAACACCCTGCTGGGC AACCCC AAGAAACGGATGCGGTACTTCGACCCCCTGCGGAACGAGTACTTCTTCGACCGGAACCGG CCCAGC TTCGACGCCATCCTGTACTACTACCAGAGCGGCGGCAGACTGCGGAGGCCCGTGAATGTG CCCCTG GACATGTTCAGCGAGGAAATCAAGTTCTACGAGCTGGGCGAGGAAGCCATGGAAAAGTTC AGAGAG GACGAGGGCTTCATCAAAGAGGAAGAGAGGCCCCTGCCCGAGAAAGAATACCAGAGACAA GTGTGG CTGCTGTTCGAGTACCCCGAGTCTAGCGGCCCTGCCAGAGTGATCGCCATCGTGTCCGTG ATGGTC ATCCTGATCTCTATCGTGATCTTCTGCCTGGAAACCCTGCCTGAGCTGAAGGACGACAAG GACTTC ACCGGCACCGTGCACCGGATCGACAACACCACCGTGATCTACAACAGCAATATCTTCACC GACCCA TTCTTCATCGTGGAAACACTGTGCATCATCTGGTTCAGCTTCGAGCTGGTCGTGCGGTTC TTCGCC TGCCCCAGCAAGACCGACTTCTTCAAGAACATCATGAACTTCATTGATATCGTGGCCATC ATCCCC TACTTCATCACCCTGGGCACCGAGATCGCCGAGCAGGAAGGCAATCAGAAGGGCGAGCAG GCCACC AGCCTGGCCATTCTGAGAGTGATCAGACTCGTGCGGGTGTTCCGGATCTTCAAGCTGAGC CGGCAC AGCAAGGGCCTGCAGATCCTGGGCCAGACACTGAAGGCCAGCATGAGAGAGCTGGGCCTG CTGATC TTCTTTCTGTTCATCGGCGTGATCCTGTTCAGCAGCGCCGTGTACTTCGCCGAGGCCGAA GAAGCC GAGAGCCACTTCAGCTCTATCCCCGACGCCTTTTGGTGGGCCGTGGTGTCCATGACCACA GTGGGC TACGGCGACATGTAnCCCGTGACAATCGGCGGCAAGATCGTGGGCAGCCTGTGTGCCATT GCCGGC GTGCTGACAGTCGCCCTGCCTGTGCCTGTGATCGTGTCCAACTTCAACTACTTCTACCAC CGGGAA ACCGAGGGGGAGGAACAGGCTCAGCTGCTGCACGTGTCCAGCCCCAATCTGGCCAGCGAC AGCGAC CTGAGCAGACGGTCTAGCAGCACCATGAGCAAGAGCGAGTACATGGAAATCGAAGAGGAC ATGAAC AACTCTATCGCCCACTACCGCCAAGTGAACATCCGGACCGCCAACTGCACCACCGCCAAC CAGAAC TGCGTGAACAAGAGCAAGCTGCTGACCGATGTCTGA wherein n is T or C Amino acid sequence of an edited human Kv1.1 comprising a valine at position 400 (underlined) (SEQ ID NO: 2)

MTVMSGENVDEASAAPGHPQDGSYPRQADHDDHECCERVVINISGLRFETQLKTLAQFPN TLLGNP KKRMRYFDPLRNEYFFDRNRPSFDAILYYYQSGGRLRRPVNVPLDMFSEEIKFYELGEEA MEKFRE DEGFIKEEERPLPEKEYQRQV LLFEYPESSGPARVIAIVSVMVILIS IVIFCLETLPELKDDKDF TGTVHRIDNTTVIYNSNIFTDPFFIVETLCII FSFELVVRFFACPSKTDFFKNIMNFIDIVAIIP YFITLGTEIAEQEGNQKGEQATSLAILRVIRLVRVFRIFKLSRHSKGLQILGQTLKASMR ELGLLI FFLFIGVILFSSAVYFAEAEEAESHFSS IPDAF AVVSMTTVGYGDMYPVTIGGKIVGSLCAIAG VLTVALPVPVIVSNFNYFYHRETEGEEQAQLLHVSSPNLASDSDLSRRSSSTMSKSEYME IEEDMN NSIAHYRQVNIRTANCTTANQNCVNKSKLLTDV

Nucleotide sequence of an exemplary human CAMK2A promoter (SEQ ID NO: 3)

TAAATAAATAAATAAATAATATAAATAATAAATGTCCAGGAATCAGAGCTCAAACTCAGA TCCTTA GTCTTAAACTCCAGTCCCTTTTCTTCCTAACTCCAAGACCTTGGAGTAAGATCTTGTGGC TGTAGG TATGGCTGATGCCCTGAAGAGTTGAAGTTGGCAGGGAAGGTGCCCAGAAAATTTTGGATT GAAGAT TTCATGGCAAGTCTCTGGCCAGTGGCCTAGCCCGGGTAAGCCATGCTATGCTCACCTCCC CACAGC CCCCTCTCGCCTTTTTTTTTTTTTTTTTTTACCTTGACTGGAAGCACAAGCAGAAACTGG GACATG AGCACCAGGAGACCAGATTTCCATGGTCCCGTTGGGGGCATGGGGTTGGGGAGAGGTTGC AGAGGA GGGCTCTGGAGGGGAGCAACTGTCACAGCTGTGAGAGGTGGGGGTGAGCAGGCAGTCAGG GCTGTT CCCTCCAGAATCCTGGGGTGTCCTCTGCACTTCTGCGCCAAGCTGGAGTGCTAGTGTGAT GGACAA GGTGGTAAGAGAGCTGAAAGAGCACGAGCATAACAAGAAAGACAGAGGCAGAAGCAAAAA AAAAAA AAAAAAAAAACAGAGGGCAACAGAGAGACAGTTACAGAGACTACAGTGATCCACAGAGGG AGAGCC ATCCCTGTGAATTAGCCATCATTTCCCTGTAAACCTTAGAACCCAGCTGTTGCCAGGGCA ACGGGG CAATACCTGTCTCTCTAGAGATGAAGTTGCCAGGGTAACTGCATCCTGTCATTCGTTCCT GGGGAC CATCCGGAATGCGGCACCCACTGGCTGTTACCATGGCAACTGCCTTTTTGCCCCACTTAA TCCCAT CCCGTCTGCTACAAGGGCCCCACAGTTGGAGGTGGGGGAGGTGGGAAGAGAAAAGATCAC TTGTGG ACAAAGTTTGCTCTATTCCACCTCCTCCAGGCCCTCCTTGGGTCCATCACCCCAGGGGTG CTGGGT CCATCCCACCCCCAGGCCCACACAGGCTTGCAGTATTGTGTGCGGTATGGTCAGGGCGTC CGAGAG CAGGTTTCGCAGTGGAAGGCAGGCAGGTGTTGGGGAGGCAGTTACCGGGGCAACGGGAAC AGGGCG TTTTGGAGGTGGTTGCCATGGGGACCTGGATGCTGACGAAGGCTCGCGAGGCTGTGAGCA GCCACA GTGCCCTGCTCAGAAGCCCCGGGCTCGTCAGTCAAACCGGTTCTCTGTTTGCACTCGGCA GCACGG GCAGGCAAGTGGTCCCTAGGTTCGGGAGCAGAGCAGCAGCGCC

Nucleotide sequence of wild-type KCNA 1 coding sequence, comprising an adenine at nucleotide position 1998 (underlined) (SEQ ID NO: 4)

ATGACGGTGATGTCTGGGGAGAACGTGGACGAGGCTTCGGCCGCCCCGGGCCACCCCCAG GATGGC AGCTACCCCCGGCAGGCCGACCACGACGACCACGAGTGCTGCGAGCGCGTGGTGATCAAC ATCTCC GGGCTGCGCTTCGAGACGCAGCTCAAGACCCTGGCGCAGTTCCCCAACACGCTGCTGGGC AACCCT AAGAAACGCATGCGCTACTTCGACCCCCTGAGGAACGAGTACTTCTTCGACCGCAACCGG CCCAGC TTCGACGCCATCCTCTACTACTACCAGTCCGGCGGCCGCCTGCGGAGGCCGGTCAACGTG CCCCTG GACATGTTCTCCGAGGAGATCAAGTTTTACGAGTTGGGCGAGGAGGCCATGGAGAAGTTC CGGGAG GACGAGGGCTTCATCAAGGAGGAGGAGCGCCCTCTGCCCGAGAAGGAGTACCAGCGCCAG GTGTGG CTGCTCTTCGAGTACCCCGAGAGCTCGGGGCCCGCCAGGGTCATCGCCATCGTCTCCGTC ATGGTC ATCCTCATCTCCATCGTCATCTTTTGCCTGGAGACGCTCCCCGAGCTGAAGGATGACAAG GACTTC ACGGGCACCGTCCACCGCATCGACAACACCACGGTCATCTACAATTCCAACATCTTCACA GACCCC TTCTTCATCGTGGAAACGCTGTGTATCATCTGGTTCTCCTTCGAGCTGGTGGTGCGCTTC TTCGCC TGCCCCAGCAAGACGGACTTCTTCAAAAACATCATGAACTTCATAGACATTGTGGCCATC ATTCCT TATTTCATCACGCTGGGCACCGAGATAGCTGAGCAGGAAGGAAACCAGAAGGGCGAGCAG GCCACC TCCCTGGCCATCCTCAGGGTCATCCGCTTGGTAAGGGTTTTTAGAATCTTCAAGCTCTCC CGCCAC TCTAAGGGCCTCCAGATCCTGGGCCAGACCCTCAAAGCTAGTATGAGAGAGCTAGGGCTG CTCATC TTTTTCCTCTTCATCGGGGTCATCCTGTTTTCTAGTGCAGTGTACTTTGCCGAGGCGGAA GAAGCT GAGTCGCACTTCTCCAGTATCCCCGATGCTTTCTGGTGGGCGGTGGTGTCCATGACCACT GTAGGA TACGGTGACATGTACCCTGTGACAATTGGAGGCAAGATCGTGGGCTCCTTGTGTGCCATC GCTGGT GTGCTAACAATTGCCCTGCCCGTACCTGTCATTGTGTCCAATTTCAACTATTTCTACCAC CGAGAA ACTGAGGGGGAAGAGCAGGCTCAGTTGCTCCACGTCAGTTCCCCTAACTTAGCCTCTGAC AGTGAC CTCAGTCGCCGCAGTTCCTCTACTATGAGCAAGTCTGAGTACATGGAGATCGAAGAGGAT ATGAAT AATAGCATAGCCCATTATAGACAGGTCAATATCAGAACTGCCAATTGCACCACTGCTAAC CAAAAC TGCGTTAATAAGAGCAAGCTACTGACCGATGTTTAA

Amino acid sequence of wild-type human Kv1.1 , comprising a isoleucine at position 400 (underlined) (SEQ ID NO: 5)

MTVMSGENVDEASAAPGHPQDGSYPRQADHDDHECCERVVINISGLRFETQLKTLAQFPN TLLGNP KKRMRYFDPLRNEYFFDRNRPSFDAILYYYQSGGRLRRPVNVPLDMFSEEIKFYELGEEA MEKFRE DEGFIKEEERPLPEKEYQRQV LLFEYPESSGPARVIAIVSVMVILIS IVIFCLETLPELKDDKDF TGTVHRIDNTTVIYNSNIFTDPFFIVETLCII FSFELVVRFFACPSKTDFFKNIMNFIDIVAIIP YF I TLGTE IAEQEGNQKGEQAT SLAI LRVI RLVRVFRI FKLSRHSKGLQI LGQTLKASMRELGLL I FFLF I GVI LF S SAVYFAEAEEAES HF S S IPDAF AVVSMTTVGYGDMYPVT I GGKIVGS LCAIAG VLTJEALPVPVIVSNFNYFYHRETEGEEQAQLLHVS SPNLASD SDLSRRS S STMSKSEYME IEEDMN NS IAHYRQVNIRTANCTTANQNCVNKSKLLTDV

Nucleotide sequence of an exemplary engineered KCNA 1 gene viral vector lacking a reporter (without bacterial plasmid portion) (SEQ ID NO: 9)

GACATTGATTATTGACTAGTTATTAATAGTAATCAATTACGGGGTCATTAGTTCATAGCC CATATA TGGAGTTCCGCGTTACATAACTTACGGTAAATGGCCCGCCTGGCTGACCGCCCAACGACC CCCGCC CATTGACGTCAATAATGACGTATGTTCCCATAGTAACGCCAATAGGGACTTTCCATTGAC GTCAAT GGGTGGAGTATTTACGGTAAACTGCCCACTTGGCAGTACATCAAGTGTATCATATGCCAA GTACGC CCCCTATTGACGTCAATGACGGTAAATGGCCCGCCTGGCATTATGCCCAGTACATGACCT TATGGG ACTTTCCTACTTGGCAGTACATCTACGTATTAGTCATCGCTATTACCATGGTGATGCGGT TTTGGC AGTACATCAATGGGCGTGGATAGCGGTTTGACTCACGGGGATTTCCAAGTCTCCACCCCA TTGACG TCAATGGGAGTTTGTTTTGGCACCAAAATCAACGGGACTTTCCAAAATGTCGTAACAACT CCGCCC CATTGACGCAAATGGGCGGTAGGCGTGTACGGTGGGAGGTCTATATAAGCAGAGCTCGTT TAGTGA ACCGGGGTCTCTCTGGTTAGACCAGATCTGAGCCTGGGAGCTCTCTGGCTAACTAGGGAA CCCACT GCTTAAGCCTCAATAAAGCTTGCCTTGAGTGCTTCAAGTAGTGTGTGCCCGTCTGTTGTG TGACTC TGGTAACTAGAGATCCCTCAGACCCTTTTAGTCAGTGTGGAAAATCTCTAGCAGTGGCGC CCGAAC AGGGACTTGAAAGCGAAAGGGAAACCAGAGGAGCTCTCTCGACGCAGGACTCGGCTTGCT GAAGCG CGCACGGCAAGAGGCGAGGGGCGGCGACTGGTGAGTACGCCAAAAATTTTGACTAGCGGA GGCTAG AAGGAGAGAGATGGGTGCGAGAGCGTCAGTATTAAGCGGGGGAGAATTAGATCGCGATGG GAAAAA ATTCGGTTAAGGCCAGGGGGAAAGAAAAAATATAAATTAAAACATATAGTATGGGCAAGC AGGGAG CTAGAACGATTCGCAGTTAATCCTGGCCTGTTAGAAACATCAGAAGGCTGTAGACAAATA CTGGGA CAGCTACAACCATCCCTTCAGACAGGATCAGAAGAACTTAGATCATTATATAATACAGTA GCAACC CTCTATTGTGTGCATCAAAGGATAGAGATAAAAGACACCAAGGAAGCTTTAGACAAGATA GAGGAA GAGCAAAACAAAAGTAAGACCACCGCACAGCAAGCGGCCGCTGATCTTCAGACCTGGAGG AGGAGA TATGAGGGACAATTGGAGAAGTGAATTATATAAATATAAAGTAGTAAAAATTGAACCATT AGGAGT AGCACCCACCAAGGCAAAGAGAAGAGTGGTGCAGAGAGAAAAAAGAGCAGTGGGAATAGG AGCTTT GTTCCTTGGGTTCTTGGGAGCAGCAGGAAGCACTATGGGCGCAGCGTCAATGACGCTGAC GGTACA GGCCAGACAATTATTGTCTGGTATAGTGCAGCAGCAGAACAATTTGCTGAGGGCTATTGA GGCGCA ACAGCATCTGTTGCAACTCACAGTCTGGGGCATCAAGCAGCTCCAGGCAAGAATCCTGGC TGTGGA AAGATACCTAAAGGATCAACAGCTCCTGGGGATTTGGGGTTGCTCTGGAAAACTCATTTG CACCAC TGCTGTGCCTTGGAATGCTAGTTGGAGTAATAAATCTCTGGAACAGATTTGGAATCACAC GACCTG GATGGAGTGGGACAGAGAAATTAACAATTACACAAGCTTAATACACTCCTTAATTGAAGA ATCGCA AAACCAGCAAGAAAAGAATGAACAAGAATTATTGGAATTAGATAAATGGGCAAGTTTGTG GAATTG GTTTAACATAACAAATTGGCTGTGGTATATAAAATTATTCATAATGATAGTAGGAGGCTT GGTAGG TTTAAGAATAGTTTTTGCTGTACTTTCTATAGTGAATAGAGTTAGGCAGGGATATTCACC ATTATC GTTTCAGACCCACCTCCCAACCCCGAGGGGACCCGACAGGCCCGAAGGAATAGAAGAAGA AGGTGG AGAGAGAGACAGAGACAGATCCATTCGATTAGTGAACGGATCTCGACGGTATCGGTTAAC TTTTAA AAGAAAAGGGGGGATTGGGGGGTACAGTGCAGGGGAAAGAATAGTAGACATAATAGCAAC AGACAT ACAAACTAAAGAATTACAAAAACAAATTACAAAATTCAAAATTTTATCGAATTCCACGGG GTTAAT CGAATAAATAAATAAATAAATAATATAAATAATAAATGTCCAGGAATCAGAGCTCAAACT CAGATC CTTAGTCTTAAACTCCAGTCCCTTTTCTTCCTAACTCCAAGACCTTGGAGTAAGATCTTG TGGCTG TAGGTATGGCTGATGCCCTGAAGAGTTGAAGTTGGCAGGGAAGGTGCCCAGAAAATTTTG GATTGA AGATTTCATGGCAAGTCTCTGGCCAGTGGCCTAGCCCGGGTAAGCCATGCTATGCTCACC TCCCCA CAGCCCCCTCTCGCCTTTTTTTTTTTTTTTTTTTACCTTGACTGGAAGCACAAGCAGAAA CTGGGA CATGAGCACCAGGAGACCAGATTTCCATGGTCCCGTTGGGGGCATGGGGTTGGGGAGAGG TTGCAG AGGAGGGCTCTGGAGGGGAGCAACTGTCACAGCTGTGAGAGGTGGGGGTGAGCAGGCAGT CAGGGC TGTTCCCTCCAGAATCCTGGGGTGTCCTCTGCACTTCTGCGCCAAGCTGGAGTGCTAGTG TGATGG ACAAGGTGGTAAGAGAGCTGAAAGAGCACGAGCATAACAAGAAAGACAGAGGCAGAAGCA AAAAAA AAAAAAAAAAAAAACAGAGGGCAACAGAGAGACAGTTACAGAGACTACAGTGATCCACAG AGGGAG AGCCATCCCTGTGAATTAGCCATCATTTCCCTGTAAACCTTAGAACCCAGCTGTTGCCAG GGCAAC GGGGCAATACCTGTCTCTCTAGAGATGAAGTTGCCAGGGTAACTGCATCCTGTCATTCGT TCCTGG GGACCATCCGGAATGCGGCACCCACTGGCTGTTACCATGGCAACTGCCTTTTTGCCCCAC TTAATC CCATCCCGTCTGCTACAAGGGCCCCACAGTTGGAGGTGGGGGAGGTGGGAAGAGAAAAGA TCACTT GTGGACAAAGTTTGCTCTATTCCACCTCCTCCAGGCCCTCCTTGGGTCCATCACCCCAGG GGTGCT GGGTCCATCCCACCCCCAGGCCCACACAGGCTTGCAGTATTGTGTGCGGTATGGTCAGGG CGTCCG AGAGCAGGTTTCGCAGTGGAAGGCAGGCAGGTGTTGGGGAGGCAGTTACCGGGGCAACGG GAACAG GGCGTTTTGGAGGTGGTTGCCATGGGGACCTGGATGCTGACGAAGGCTCGCGAGGCTGTG AGCAGC CACAGTGCCCTGCTCAGAAGCCCCGGGCTCGTCAGTCAAACCGGTTCTCTGTTTGCACTC GGCAGC ACGGGCAGGCAAGTGGTCCCTAGGTTCGGGAGCAGAGCAGCAGCGCCGGATCCGCCACCA TGACCG TGATGAGCGGCGAGAACGTGGACGAGGCCTCTGCCGCTCCTGGACACCCTCAGGATGGCA GCTATC CCAGACAGGCCGACCACGACGATCACGAGTGCTGCGAGCGGGTCGTGATCAACATCAGCG GCCTGA GATTCGAGACACAGCTGAAAACCCTGGCCCAGTTCCCCAACACCCTGCTGGGCAACCCCA AGAAAC GGATGCGGTACTTCGACCCCCTGCGGAACGAGTACTTCTTCGACCGGAACCGGCCCAGCT TCGACG CCATCCTGTACTACTACCAGAGCGGCGGCAGACTGCGGAGGCCCGTGAATGTGCCCCTGG ACATGT TCAGCGAGGAAATCAAGTTCTACGAGCTGGGCGAGGAAGCCATGGAAAAGTTCAGAGAGG ACGAGG GCTTCATCAAAGAGGAAGAGAGGCCCCTGCCCGAGAAAGAATACCAGAGACAAGTGTGGC TGCTGT TCGAGTACCCCGAGTCTAGCGGCCCTGCCAGAGTGATCGCCATCGTGTCCGTGATGGTCA TCCTGA TCTCTATCGTGATCTTCTGCCTGGAAACCCTGCCTGAGCTGAAGGACGACAAGGACTTCA CCGGCA CCGTGCACCGGATCGACAACACCACCGTGATCTACAACAGCAATATCTTCACCGACCCAT TCTTCA TCGTGGAAACACTGTGCATCATCTGGTTCAGCTTCGAGCTGGTCGTGCGGTTCTTCGCCT GCCCCA GCAAGACCGACTTCTTCAAGAACATCATGAACTTCATTGATATCGTGGCCATCATCCCCT ACTTCA TCACCCTGGGCACCGAGATCGCCGAGCAGGAAGGCAATCAGAAGGGCGAGCAGGCCACCA GCCTGG CCATTCTGAGAGTGATCAGACTCGTGCGGGTGTTCCGGATCTTCAAGCTGAGCCGGCACA GCAAGG GCCTGCAGATCCTGGGCCAGACACTGAAGGCCAGCATGAGAGAGCTGGGCCTGCTGATCT TCTTTC TGTTCATCGGCGTGATCCTGTTCAGCAGCGCCGTGTACTTCGCCGAGGCCGAAGAAGCCG AGAGCC ACTTCAGCTCTATCCCCGACGCCTTTTGGTGGGCCGTGGTGTCCATGACCACAGTGGGCT ACGGCG ACATGTAnCCCGTGACAATCGGCGGCAAGATCGTGGGCAGCCTGTGTGCCATTGCCGGCG TGCTGA CAGTCGCCCTGCCTGTGCCTGTGATCGTGTCCAACTTCAACTACTTCTACCACCGGGAAA CCGAGG GGGAGGAACAGGCTCAGCTGCTGCACGTGTCCAGCCCCAATCTGGCCAGCGACAGCGACC TGAGCA GACGGTCTAGCAGCACCATGAGCAAGAGCGAGTACATGGAAATCGAAGAGGACATGAACA ACTCTA TCGCCCACTACCGCCAAGTGAACATCCGGACCGCCAACTGCACCACCGCCAACCAGAACT GCGTGA ACAAGAGCAAGCTGCTGACCGATGTCTGAGTCGACAATCAACCTCTGGATTACAAAATTT GTGAAA GATTGACTGGTATTCTTAACTATGTTGCTCCTTTTACGCTATGTGGATACGCTGCTTTAA TGCCTT TGTATCATGCTATTGCTTCCCGTATGGCTTTCATTTTCTCCTCCTTGTATAAATCCTGGT TGCTGT CTCTTTATGAGGAGTTGTGGCCCGTTGTCAGGCAACGTGGCGTGGTGTGCACTGTGTTTG CTGACG CAACCCCCACTGGTTGGGGCATTGCCACCACCTGTCAGCTCCTTTCCGGGACTTTCGCTT TCCCCC TCCCTATTGCCACGGCGGAACTCATCGCCGCCTGCCTTGCCCGCTGCTGGACAGGGGCTC GGCTGT TGGGCACTGACAATTCCGTGGTGTTGTCGGGGAAATCATCGTCCTTTCCTTGGCTGCTCG CCTGTG TTGCCACCTGGATTCTGCGCGGGACGTCCTTCTGCTACGTCCCTTCGGCCCTCAATCCAG CGGACC TTCCTTCCCGCGGCCTGCTGCCGGCTCTGCGGCCTCTTCCGCGTCTTCGCCTTCGCCCTC AGACGA GTCGGATCTCCCTTTGGGCCGCCTCCCCGCCTGGAATTCGAGCTCGGTACCTTTAAGACC AATGAC TTACAAGGCAGCTGTAGATCTTAGCCACTTTTTAAAAGAAAAGGGGGGACTGGAAGGGCT AATTCA CTCCCAACGAAGACAAGATCTGCTTTTTGCTTGTACTGGGTCTCTCTGGTTAGACCAGAT CTGAGC CTGGGAGCTCTCTGGCTAACTAGGGAACCCACTGCTTAAGCCTCAATAAAGCTTGCCTTG AGTGCT TCAAGTAGTGTGTGCCCGTCTGTTGTGTGACTCTGGTAACTAGAGATCCCTCAGACCCTT TTAGTC AGTGTGGAAAATCTCTAGCA wherein n is T or C

Amino acid sequence of an edited human Kv1.1 comprising a valine at position 400 (underlined) and a valine at position 379 substitution (bolded) (SEQ ID NO: 14)

MTVMSGENVDEASAAPGHPQDGSYPRQADHDDHECCERVVINISGLRFETQLKTLAQFPN TLLGNP KKRMRYFDPLRNEYFFDRNRPSFDAILYYYQSGGRLRRPVNVPLDMFSEEIKFYELGEEA MEKFRE DEGFIKEEERPLPEKEYQRQV LLFEYPESSGPARVIAIVSVMVILIS IVIFCLETLPELKDDKDF TGTVHRIDNTTVIYNSNIFTDPFFIVETLCII FSFELVVRFFACPSKTDFFKNIMNFIDIVAIIP YFITLGTEIAEQEGNQKGEQATSLAILRVIRLVRVFRIFKLSRHSKGLQILGQTLKASMR ELGLLI FFLFIGVILFSSAVYFAEAEEAESHFSS IPDAF AVVSMTTVGYGDMVPVTIGGKIVGSLCAIAG VLTVALPVPVIVSNFNYFYHRETEGEEQAQLLHVSSPNLASDSDLSRRSSSTMSKSEYME IEEDMN NSIAHYRQVNIRTANCTTANQNCVNKSKLLTDV