Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
OPTIMIZED LENTIVIRAL VECTOR FOR STEM CELL GENE THERAPY OF HEMOGLOBINOPATHIES
Document Type and Number:
WIPO Patent Application WO/2018/106724
Kind Code:
A1
Abstract:
In certain embodiments an optimized derivative of the CCLc-βAS3-FB lentiviral vector termed (CCLc-mGata/ANK-CoreLCR-βAS3-FB), is provided which is capable of driving lineage-restricted expression of a beta-globin gene (e.g., an anti-sickling β-globin like gene (βΑ83)). In certain embodiments the vectors described herein comprise novel defined LCR HS core sequences (HS2(~420 bp), HS3(~340 bp), HS4(~410 bp)) which can be used to replace the putative LCR HS sequences present within the "mini-LCR" (~3.6 kb reduced to ~1.2 kb) to produce an "optimized mini-LCR".

Inventors:
KOHN DONALD B (US)
MORGAN RICHARD A (US)
HOLLIS ROGER P (US)
Application Number:
PCT/US2017/064766
Publication Date:
June 14, 2018
Filing Date:
December 05, 2017
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
UNIV CALIFORNIA (US)
International Classes:
C12N15/11; C12N15/12; C12N15/63; C12N15/85; C12N15/867
Domestic Patent References:
WO2016037138A12016-03-10
WO2013173599A12013-11-21
WO2016118715A12016-07-28
WO2016037138A12016-03-10
WO2014043131A12014-03-20
Foreign References:
US20150224209A12015-08-13
US20080069805A12008-03-20
US5994136A1999-11-30
US20030039636A12003-02-27
US6136597A2000-10-24
US5686279A1997-11-11
US4522811A1985-06-11
US20150224209A12015-08-13
Other References:
HOBAN ET AL.: "Genetic treatment of a molecular disorder: gene therapy approaches to sickle cell disease", BLOOD, vol. 127, no. 7, 12 January 2016 (2016-01-12), pages 839 - 848, XP055510604
ROMERO ET AL.: "P-globin gene transfer to human bone marrow for sickle cell disease", THE JOURNAL OF CLINICAL INVESTIGATION, vol. 123, no. 8, 1 August 2013 (2013-08-01), pages 3317 - 3330, XP055243792
BOLANOS-MEADEBRODSKY, CURR. OPIN. ONCOL., vol. 21, no. 2, 2009, pages 158 - 161
VOSKARIDOU ET AL., BLOOD, vol. 115, no. 12, 2010, pages 2354 - 2363
EATONHOFRICHTER, BLOOD, vol. 70, no. 5, 1987, pages 1245 - 1266
ADLER ET AL., BLOOD, vol. 97, no. 10, 2001, pages 3313 - 3314
REES, LANCET, vol. 376, no. 9757, 2010, pages 2018 - 2031
SHENOY, HEMATOLOGY AM SOC HEMATOL EDUC PROGRAM., 2011, pages 273 - 279
KAMANI ET AL., BIOL. BLOOD MARROW TRANSPLANT., vol. 18, no. 8, 2012, pages 1265 - 1272
LOCATELLIPAGLIARA, PEDIATR. BLOOD CANCER., vol. 59, no. 2, 2012, pages 372 - 376
ABBOUD ET AL., LANCET, vol. 351, no. 9107, 1998, pages 959
FITZHUGH ET AL., CYTOTHERAPY, vol. 11, no. 4, 2009, pages 464 - 471
NEUMAYR ET AL., AM. J. HEMATOL., vol. 57, no. 2, 1998, pages 101 - 108
LISOWSKISADELAIN, BR. J. HAEMATOL., vol. 141, no. 3, 2008, pages 335 - 345
GELINAS ET AL., ADV. EXP. MED. BIOL., vol. 271, 1989, pages 135 - 148
GELINAS ET AL., PROG. CLIN. BIOL. RES., vol. 316B, 1989, pages 235 - 249
MAY ET AL., NATURE, vol. 406, no. 6791, 2000, pages 82 - 86
PAWLIUK ET AL., SCIENCE, vol. 294, no. 5550, 2001, pages 2368 - 2371
LEVASSEUR ET AL., BLOOD, vol. 102, no. 13, 2003, pages 4312 - 4319
PUTHENVEETIL ET AL., BLOOD, vol. 104, no. 12, 2004, pages 3445 - 3453
MICCIO ET AL., PROC. NATL. ACAD. SCI. USA, vol. 105, no. 30, 2008, pages 10547 - 10552
PESTINA ET AL., MOL. THER., vol. 17, no. 2, 2008, pages 245 - 252
PLATT, N. ENGL. J. MED., vol. 330, no. 23, 1994, pages 1639 - 1644
CHAKALOVA ET AL., BLOOD, vol. 105, no. 5, 2005, pages 2154 - 2160
EUR. J. HAEMATOL., vol. 79, no. 6, 2007, pages 516 - 525
PERSONS ET AL., BLOOD, vol. 101, no. 6, 2002, pages 2175 - 2183
PERUMBETI, BLOOD, vol. 114, no. 6, 2009, pages 1174 - 1185
BREDA ET AL., PLOS ONE, vol. 7, no. 3, 2012, pages e32345
NAGEL ET AL., PROC. NATL. ACAD. SCI., USA, vol. 76, no. 2, 1979, pages 670 - 672
CAVAZZANA-CALVO ET AL., NATURE, vol. 467, no. 7313, 2010, pages 318 - 322
MCCUNE ET AL., PROC. NATL. ACAD. SCI. USA, vol. 91, no. 21, 1994, pages 3054 - 9856
LEVASSEUR ET AL., J. BIOL. CHEM., vol. 279, no. 26, 2004, pages 27518 - 27524
FORRESTER ET AL., PROC. NATL. ACAD. SCI. USA, vol. 83, 1986, pages 1359 - 1363
CARILLO ET AL., APPL. MATH, vol. 48, 1988, pages 1073
"Biocomputing: Informatics and Genome Projects", 1993, ACADEMIC PRESS
"Computer Analysis of Sequence Data", 1994, ACADEMIC PRESS
"Sequence Analysis in Molecular Biology", 1987, ACADEMIC PRESS
"Sequence Analysis Primer", 1991, STOCKTON PRESS
NEEDLEMANWUNSCH, J. MOL. BIOL., vol. 48, 1970, pages 443 - 453
SMITHWATERMAN, ADV. APPL. MATH., vol. 2, 1981, pages 482 - 489
SMITH ET AL., NUCLEIC ACIDS RES., vol. 11, 1983, pages 2205 - 2220
ALTSCHUL ET AL., J. MOL. BIOL., vol. 215, 1990, pages 403 - 410
ANTONIOU ET AL., NUCL. ACIDS RES., vol. 26, no. 3, 1998, pages 721 - 729
ZUFFEREY ET AL., J VIROL., vol. 72, no. 11, 1998, pages 8463 - 8471
BURGESS-BEUSSE ET AL.: "99", PROC. NATL. ACAD. SCI. USA, 2002, pages 16433
ZHAN, HUM. GENET., vol. 109, 2001, pages 471
PRIOLEAU ET AL., EMBO J., vol. 18, 1999, pages 4035 - 4048
CHUNG ET AL., CELL, vol. 98, 1999, pages 387 - 396
PIKAART ET AL., GENES DEV., vol. 12, 1998, pages 2852 - 2862
WANG ET AL., NAT. BIOTECHNOL., vol. 15, 1997, pages 239 - 243
TABOIT-DAMERON ET AL., TRANSGENIC RES., vol. 8, 1999, pages 223 - 235
CHUNG ET AL., PROC. NATL. ACAD. SCI., USA, vol. 94, 1997, pages 575 - 580
RAMEZANI ET AL., STEM CELL, vol. 26, 2008, pages 3257 - 3266
"Genbank", Database accession no. AF003887
ROMERO ET AL., J. CLIN. INVEST., vol. 128, no. 8, 2013, pages 3317 - 3330
See also references of EP 3548617A4
Attorney, Agent or Firm:
HUNTER, Tom et al. (US)
Download PDF:
Claims:
CLAIMS

What is claimed is;

1. A recombinant lentiviral vector (LV) comprising:

an expression cassette comprising a nucleic acid construct comprising: a human β-globin locus control region comprising at least one hypersensitive site (HS) core sequence from the group consisting of HS2 (~420bp), HS3 (~ 340 bp), and HS4 (~410bp); and

a recombinant human beta globin gene encoding a beta globin polypeptide; and

where said LV is a TAT-independent and self-inactivating (SIN) lentiviral vector.

2. The vector of claim 1 , wherein said β-globin locus control region comprises hypersensitive site (HS) core sequence HS2.

3. The vector of claim 1, wherein said β-globin locus control region comprises hypersensitive site (HS) core sequence HS3.

4. The vector of claim 1, wherein said β-globin locus control region comprises hypersensitive site (HS) core sequence HS4.

5. The vector of claim 1, wherein said β-globin locus control region comprises hypersensitive site (HS) core sequences HS2 and HS3. 6. The vector of claim 1 , wherein said β-globin locus control region comprises hypersensitive site (HS) core sequences HS2 and HS4.

7. The vector of claim 1 , wherein said β-globin locus control region comprises hypersensitive site (HS) core sequences HS3 and HS4.

8. The vector of claim 1, wherein said β-globin locus control region comprises hypersensitive site (HS) core sequences HS2, HS3, and HS4.

9. The vector according to any one of claims 1-8, wherein said HS2 core sequence consists of a nucleic acid sequence having at least 90%, or at least 95%, or at least 98% sequence identity with the sequence tanging from S484 to S908 of SEQ ID NO:l, or from 5077 to 5501 of SEQ ID NO:2.

10. The vector of claim 9, wherein said HS2 core sequence consists of a nucleic acid sequence ranging from 5484 to 5908 of SEQ ID NO:l, or from 5077 to 5501 ofSEQ ID NO:2.

11. The vector according to any one of claims 1-10, wherein said HS3 core sequence consists of a nucleic acid sequence having at least 90%, or at least 95%, or at least 98% sequence identity with the sequence ranging from 5909 to 6252 of SEQ ID NO: 1 , or from 5502 to 5845 of SEQ ID NO:2. 12. The vector of claim 11 , wherein said HS3 core sequence consists of a nucleic acid sequence ranging from 5909 to 6252 of SEQ ID NO:l, or from 5502 to 5845 of SEQ IDNO:2.

13. The vector according to any one of claims 1-12, wherein said HS4 core sequence consists of a nucleic acid sequence having at least 90%, or at least 95%, or at least 98% sequence identity with the sequence ranging from 6253 to 6662 of SEQ ID NO: 1 , or from 5846 to 6255 of SEQ ID NO:2.

14. The vector of claim 13, wherein said HS4 core sequence consists of a nucleic acid sequence ranging from 6253 to 6662 of SEQ ID NO:l, or from 5846 to 6255 of SEQ IDNO:2. 15. The vector according to any one of claims 1-8, wherein said human β-globin locus control region comprises the nucleic acid sequence ranging from 5484 to 6662 of SEQ ID NO:l, or from 5077 to 6255 of SEQ ID NO:2.

16. The vector of claim 15, wherein said human β-globin locus control region consists of the nucleic acid sequence ranging from 5484 to 6662 of SEQ ID NO: 1, or from 5077 to 6255 of SEQ ID NO:2.

17. The vector according to any one of claims 1-16, wherein said human beta globin gene comprises a wild-type beta globin gene.

18. The vector according to any one of claims 1-16, wherein said human beta globin gene comprises an anti- sickling human beta globin gene encoding an anti- sickling beta globin polypeptide.

19. The vector of claim 18, wherein said anti-sickling human beta globin gene encoding an anti-sickling-beta globin polypeptide comprise one or more mutations selected from the group consistnng of Glyl6Asp, Glu22Ala and Thr87Gln.

20. The vector of claim 19, wherein said beta globin gene comprises the mutation Glyl6Asp.

21. The vector according to any one of claims 19-20, wherein said beta globin gene comprises the mutation Glu22Ala.

22. The vector according to any one of claims 19-21, wherein said beta globin gene comprises the mutation Thr87Gln.

23. The vector of claim 19, wherein said anti-sickling human β-globin gene comprises about 2.3 kb of recombinant human β-globin gene including exons and introns under the control of the human β -globin gene 5 ' promoter and the human β-globin 3' enhancer.

24. The vector according to any one of claims 1-23, wherein said β- globin gene comprises β-globin intron 2 with a 37S bp Rsal deletion from IVS2.

25. The vector according to any one of claims 1-24, wherein said β- globin gene comprises an Sspl (S) to Rsal (R) deletion (~220bp).

26. The vector according to any one of claims 1 -25, wherein said vector comprises a human Ankyrin insulator element.

27. The vector of claim 26, wherein said human Ankyrin insulator element comprises a nucleic acid sequence having at least 90%, or at least 95%, or at least 98% sequence identity with the sequence ranging from 5325 to 5483 of SEQ ID NO: 1 , or from 6458 to 6616 of SEQ JD NO:2.

28. The vector of claim 27, wherein said human Ankyrin insulator element consists of a nucleic acid sequence ranging from 5325 to 5483 of SEQ ID NO:l, or from 6458 to 6616 of SEQ ID NO:2.

29. The vector according to any one of claims 26-28, wherein said human ankyrin insulator is adjacent to HS4.

30. The vector according to any one of claims 26-28, wherein said human ankyrin insulator is adjacent to HS2. 31. The vector according to any one of claims 1 -30, wherein said vector comprises a murine GATA1-HS2.

32. The vector of claim 31, wherein said murine GATA1-HS2 comprises a nucleic acid sequence having at least 90%, or at least 95%, or at least 98% sequence identity with the sequence ranging from 6256 to 6457 SEQ ID NO:2. 33. The vector of claim 32, wherein said murine GATA1-HS2 consists of the nucleic acid sequence ranging from 6256 to 6457 SEQ ID NO:2.

34. The vector according to any one of claims 31-33, wherein said GATA1-HS2 is adjacent to HS2.

35. The vector according to any one of claims 31-33, wherein said GATA1-HS2 is adjacent to HS4.

36. The vector according to any one of claims 1-35, further comprising an insulator in the 3' LTR.

37. The vector of claim 36, wherein said insulator comprises FB

(FII/BEAD-A), a 77 bp insulator element, which contains the minimal CTCF binding site enhancer-blocking components of the chicken β-globin 5' Dnasel-hypersensitive site 4 (5' HS4).

38. The vector according to any one of claims 1-37, wherein said vector comprises a ψ region vector genome packaging signal.

39. The vector according to any one of claims 1-38, wherein the 5' LTR comprises a CMV enhancer/promoter.

40. The vector according to any one of claims 1-39, wherein said vector comprises a Rev Responsive Element (RRE).

41. The vector according to any one of claims 1-40, wherein said vector comprises a central polypurine tract

42. The vector according to any one of claims 1-41, wherein said vector comprises apost-translational regulatory element. 43. The vector of claim 42, wherein the posttranscriptional regulatory element is modified Woodchuck Post-transcriptional Regulatory Element (WPRE).

44. The vector of claim 1 , wherein said vector comprises the nucleic acid sequence of SEQ ID NO: 1.

45. The vector of claim 1, wherein said vector comprises the nucleic acid sequence of SEQ ID NO:2.

46. The vector according to any one of claims 1-45, wherein said vector is incapable of reconstituting a wild-type lentivims through recombination.

47. A host cell transduced with a vector according to any one of claims 1-

46. 48. The host cell of claim 47, wherein the cell is a stem cell.

49. The host cell of claim 48, wherein said cell is a stem cell derived from bone marrow, and/or from umbilical cord blood, and/or from peripheral blood.

50. The host cell of claim 47, wherein the cell is a 293T cell.

51. The host cell of claim 47, wherein, wherein the cell is a human hematopoietic progenitor cell.

52. The host cell of claim 51 , wherein the human hematopoietic progenitor cell is a CD34+ cell.

53. A method of treating a hemoglobinopathy, in a subject, said method comprising:

transducing a stem cell and/or progenitor cell from said subject with a vector according to any one of claims 1-46; and transplanting said transduced cell or cells derived therefrom into said subject where said cells or derivatives therefrom express said anti- sickling human beta globin gene.

54. The method of claim 53, wherein the cell is a stem cell. 55. The host cell of claim 53, wherein said cell is a stem cell derived from bone marrow.

56. The method of claim 53, wherein, wherein the cell is a human hematopoietic progenitor cell.

57. The method of claim 56, wherein the human hematopoietic progenitor cell is a CD34+ cell .

58. The method according to any one of claims 53-57, wherein said hemoglobinopathy is sickle cell disease.

59. The method according to any one of claims 53-57, wherein said hemoglobinopathy is β-thalassemia.

Description:
OPTIMIZED LENTIVIRAL VECTOR FOR STEM CELL GENE THERAPY OF HEMOGLOBINOPATHIES

CROSS-REFERENCE TO RELATED APPLICATIONS

[0001] This application claims benefit of and priority to USSN 62/430,157, filed on December 5, 2016, which is incorporated herein by reference in its entirey for all purposes.

STATEMENT OF GOVERNMENTAL SUPPORT

[ Not Applicable ]

BACKGROUND

[0002] Sickle cell disease (SCD) is one of the most common monogenic disorders worldwide and is a major cause of morbidity and early mortality (Hoffman et al. (2009) Hematology: Basic Principles and Practice. 5th ed. London, United Kingdom, Churchill Livingstone). SCD affects approximately 80,000 Americans, and causes significant neurologic, pulmonary, and renal injury, as well as severe acute and chronic pain that adversely impacts quality of life. It is estimated that approximately 240,000 children are born annually in Africa with SCD and 80% die by their second birthday. The average lifespan of subjects with SCD in the United States is approximately 40 years and this has remained unchanged over the last 3-4 decades.

[0003] SCD is caused by a single amino acid change in β-globin (Glu 6 to Val 6) which leads to hemoglobin polymerization and red blood cell (rbc) sickling. SCD typically results in continual low-grade ischemia and episodic exacerbations or "crises" resulting in tissue ischemia, organ damage, and premature death.

[0004] Although SCD is well characterized, there is still no ideal long-term treatment. Current therapies are based on induction of fetal hemoglobin (HbF) to inhibit polymerization of sickle hemoglobin (HbS) (Voskaridou et al. (2010) Blood, 115(12): 2354-2363) and cell dehydration (Eaton and Hofrichter (1987) Blood, 70(5): 1245-1266) or reduction of the percentage of HbS by transfusions (Stamatoyannopoulos et al, eds. (2001) Molecular Basis of Blood Diseases. 3rd ed. Philadelphia, Pennsylvania, USA: WB

Saunders). Allogeneic human stem cell transplantation (HSCT) from bone marrow (BM) or umbilical cord blood (UCB) or mobilized peripheral blood stem cells (mPBSC) is a potentially curative therapy, although only a small percentage of patients have undergone this procedure, mostly children with severe symptoms who had HLA-matched sibling donors (Bolanos-Meade andBrodsky (2009) Curr. Opin. Oncol. 21(2): 158-161; Rees etal (2010) Lancet, 376(9757): 2018-2031; Shcnoy (2011) Hematology Am Soc Hematol Educ Program.2011 : 273-279).

[0005] Transplantation of allogeneic cells carries the risk of graft-versus host disease (GvHD), which can be a cause of extensive morbidity. HSCT using UCB from matched unrelated donors holds reduced risk of acute or chronic GvHD compared with using BM; however, there is a higher probability of engraftment failure using UCB as a result of its lower cell dose and immunologic immaturity (Kamani et al. (2012) Biol. Blood Marrow Transplant. 18(8): 1265-1272; Locatelli and Pagliara (2012) Pediatr. Blood Cancer. 59(2): 372-376).

[0006] Gene therapy with autologous human stem cells (HSCs) is an alternative to allogeneic HSCT, since it avoids the limitations of finding a matched donor and the risks of GvHD and graft rejection. For gene therapy application in SCD patients, the safest source for autologous HSC would be BM, due to the complications previously described when G- CSF was used to collect autologous peripheral blood stem cells (PBSCs) in SCD patients (Abboud et al. (1998) Lancei351(9107): 959; Adler et al. (2001) Blood, 97(10): 3313-3314; Fitzhugh et al. (2009) Cytotherapy, 11(4): 464-471). Although general anesthesia imposes a risk for SCD patients as well, current best medical practices can minimize these (Neumayr etal (1998) Am. J. Hematol 57(2): 101-108).

[0007] The development of integrating vectors for β-globin gene transfer has been challenging due to the complex regulatory elements needed for high-level, eryt hroid- specific expression (Lisowski and Sadelain (2008) Br. J. Haematol. 141(3): 335-345). γ- Retroviral vectors were unable to transfer these β-globin expression cassettes intact (Gelinas etal (1989) Adv. Exp. Med. Biol. 271: 135-148; Gelinas etal. (1989) Prog. Clin. Biol. Res. 316B: 235-249). In contrast, lentiviral vectors (LV) can transfer β-globin cassettes intact with relatively high efficiency, although the titers of these vectors are reduced compared with those of vectors bearing simpler cassettes (May et al. (2000) Nature 406(6791): 82-86; Pawliuk et al (2001) Science, 294(5550): 2368-2371). In the last decade, many groups have developed different β-globin LV for targeting β-hemoglobinopathies, with successful therapeutic results following transplantation of ex vivo- modified HSC in mouse models (May et al. (2000) Nature 406(6791): 82-86; Pawliuk et al. (2001) Science, 294(5550): 2368-2371; Levasseur et al. (2003) Blood, 102(13):4312-4319; Hanawa etal. (2004) Blood, 104(8): 2281-2290; Puthenveetil et al. (2004) Blood, 104(12): 3445-3453; Miccio et al (2008) Proc. Natl. Acad. Sci. USA, 105(30): 10547- 10552; Pestina ef a/. (2008) Mo/. Ther. 17(2): 245-252).

[0008] Sickle patients with hereditary persistence of fetal hemoglobin (HbF)

(HPFH) have improved survival and amelioration of clinical symptoms, with maximal clinical benefits observed when the HbF is elevated above threshold values (e.g., 8%-15% of the total cellular Hb) (Voskaridou et al. (2010) Blood, 115(12): 2354-2363; Piatt et al. (1994) N. Engl. J. Med. 330(23): 1639-1644). Therefore, some gene therapy strategies have employed viral vectors carrying the human γ-globin gene (HBG1/2). However, these constructs expressed HbF poorly in adult erythroid cells, since fetal-specific transcription factors are required for high-level expression of the γ-globin gene (Chakalova et al. (2005) Bloodl05(5): 2154-2160; Russell (2007) Eur. J. Haematol. 79(6): 516-525). These limitations have been overcome by embedding the exons encoding human γ-globin within the human β-globin gene 5' promoter and 3' enhancer elements (Hanawa et al. (2004) Blood, 104(8): 2281-2290; Persons etal. (2002) Blood, 101(6): 2175-2183; Penimbeti etal. (2009) Blood, 114(6): 1174-1185). Breda et al. (2012) PLoS One, 7(3): e32345 used an LV vector encoding the human hemoglobin (HBB) gene to increase the expression of normal HbA in CD34 + -derived erythroid cells from SCD patients, however, the expression level needed when the HBB gene is used would be higher than would be required for HBG1/2 gene expression to achieve therapeutic benefits in SCD patients.

[0009] Another approach is to modify β-globin genes to confer antisickling activity by substituting key amino acids from γ-globin. The modified β-globin cassette should yield the necessary high-level, erythroid-specific expression in adult erythroid cells. Pawliuk et al. (2001) Science, 294(5550): 2368-2371 designed an LV carrying a human β-globin gene with the amino acid modification T87Q. The glutamine at position 87 of γ-globin has been implicated in the anti-sickling activity of HbF (Nagel et al. (1979) Proc. Natl. Acad. ScL, USA, 76(2): 670-672). This anti-sickling construct corrected SCD in 2 murine models of the disease, and a similar LV has been used in a clinical trial for β-thalassemia and SCD in France (Cavazzana-Calvo et al. (2010) Nature, 467(7313): 318-322).

[0010] Townes and colleagues have taken a similar approach, developing a recombinant human anti-sickling β-globin gene (HBBAS3) encoding a β-globin protein (HbAS3) that has 3 amino substitutions compared with the original (HbA): T87Q for blocking the lateral contact with the canonical Val 6 of HbS, E22A to disrupt axial contacts (McCune et al. (1994) Proc. Natl. Acad. Sci. USA, 91(21): 9852-9856) and G16D, which confers a competitive advantage over sickle-P-globin chains for interaction with the <x- globin polypeptide. Functional analysis of the purified HbAS3 protein demonstrated that this recombinant protein had potent activity to inhibit HbS tetramer polymerization (Levasseur et al. (2004) J. Biol Chem. 279(26): 27518-27524.). Levasseur et al. (2003) Blood, 102(13): 4312-4319, showed efficient transduction of BM stem cells from a murine model of SCD with a self-inactivating (SIN) LV carrying the HBBAS3 transgene that resulted in normalized rbc physiology and prevented the pathological manifestations of SCD.

[0011] Unfortunately, current β-globin expression vectors, suffer from low vector titer and sub-optimal gene transfer to hematopoietic stem cells, representing a major barrier toward the effective implementation of this gene therapy strategy to the clinic.

SUMMARY

[0012] Various embodiments cnetemplated herein may include, but need not be limited to, one or more of the following:

[0013] Embodiment 1 :: A recombinant lenti viral vector (LV) comprising:

[0014] an expression cassette comprising a nucleic acid construct comprising:

[0015] a human β-globin locus control region comprising at least one hypersensitive site (HS) core sequence from the group consisting of HS2 (e.g., an HS2 consisting of an HS2 core sequence, ~420bp), HS3 (e.g., an HS3 consisting of an HS3 core sequence, ~ 340 bp), and HS4 (e.g., an HS4 consisting of an HS4 core sequence, ~410bp); and

[0016] a recombinant human beta globin gene encoding a beta globin polypeptide; and where said LV is a TAT-independent and self-inactivating (SIN) lenti viral vector.

[0017] Embodiment 2: The vector of embodiment 1, wherein said β-globin locus control region comprises hypersensitive site (HS) core sequence HS2 (e.g., an HS2 consisting of an HS2 core sequence).

[0018] Embodiment 3: The vector of embodiment 1, wherein said β-globin locus control region comprises hypersensitive site (HS) core sequence HS3 (e.g., an HS3 consisting of an HS3 core sequence). [0019] Embodiment 4: The vector of embodiment 1 , wherein said β-globin locus control region comprises hypersensitive site (HS) core sequence HS4 (e.g„ an HS4 consisting of an HS4 core sequence).

[0020] Embodiment 5: The vector of embodiment 1, wherein said β-globin locus control region comprises hypersensitive site (HS) core sequences HS2 and HS3 (e.g., an HS2 consisting of an HS2 core sequence and an HS3 consisting of an HS3 core sequence).

[0021] Embodiment 6: The vector of embodiment 1, wherein said β-globin locus control region comprises hypersensitive site (HS) core sequences HS2 and HS4 (e.g., an HS2 consisting of an HS2 core sequence and an HS4 consisting of an HS4 core sequence).

[0022] Embodiment 7: The vector of embodiment 1, wherein said β-globin locus control region comprises hypersensitive site (HS) core sequences HS3 and HS4 (e.g., an HS3 consisting of an HS3 core sequence and an HS4 consisting of an HS4 core sequence).

[0023] Embodiment 8: The vector of embodiment 1, wherein said β-globin locus control region comprises hypersensitive site (HS) core sequences HS2, HS3, and HS4(e.g., an HS2 consisting of an HS2 core sequence, an HS3 consisting of an HS3 core sequence, and an HS4 consisting of an HS4 core sequence).

[0024] Embodiment 9: The vector according to any one of embodiments 1-8, wherein said HS2 core sequence consists of a nucleic acid sequence having at least 90%, or at least 95%, or at least 98% sequence identity with the sequence ranging from 5484 to 5908 of SEQ ID NO: 1, or from 5077 to 5501 of SEQ ID NO:2.

[0025] Embodiment 10: The vector of embodiment 9, wherein said HS2 core sequence consists of a nucleic acid sequence ranging from 5484 to 5908 of SEQ ID NO:l, or from 5077 to 5501 of SEQ ID NO:2.

[0026] Embodiment 11 : The vector according to any one of embodiments 1-10, wherein said HS3 core sequence consists of a nucleic acid sequence having at least 90%, or at least 95%, or at least 98% sequence identity with the sequence ranging from 5909 to 6252 of SEQ ID NO: 1, or from 5502 to 5845 of SEQ ID NO:2.

[0027] Embodiment 12: The vector of embodiment 11 , wherein said HS3 core sequence consists of a nucleic acid sequence ranging from 5909 to 6252 of SEQ ID NO:l, or from 5502 to 5845 of SEQ ID NO:2.

[0028] Embodiment 13: The vector according to any one of embodiments 1-12, wherein said HS4 core sequence consists of a nucleic acid sequence having at least 90%, or at least 95%, or at least 98% sequence identity with the sequence ranging from 6253 to 6662 of SEQ ID NO: 1, or from 5846 to 6255 of SEQ ID NO:2.

[0029] Embodiment 14: The vector of embodiment 13, wherein said HS4 core sequence consists of a nucleic acid sequence ranging from 6253 to 6662 of SEQ ID NO: 1 , or from 5846 to 6255 of SEQ ID NO:2.

[0030] Embodiment 15: The vector according to any one of embodiments 1-8, wherein said human β-globin locus control region comprises the nucleic acid sequence ranging from 5484 to 6662 of SEQ ID NO:l, or from 5077 to 6255 of SEQ ID NO:2.

[0031] Embodiment 16: The vector of embodiment 15, wherein said human β- globin locus control region consists of the nucleic acid sequence ranging from 5484 to 6662 of SEQ ID NO: 1, or from 5077 to 6255 of SEQ ID NO:2.

[0032] Embodiment 17: The vector according to any one of embodiments 1-16, wherein said human beta globin gene comprises a wild-type beta globin gene and/or a wild- type gamma globin gene.

[0033] Embodiment 18: The vector according to any one of embodiments 1-16, wherein said human beta globin gene comprises an anti- sickling human beta globin gene encoding an anti-sickling beta globin polypeptide.

[0034] Embodiment 19: The vector of embodiment 18, wherein said anti-sickling human beta globin gene encoding an anti-sickling-beta globin polypeptide comprise one or more mutations selected from the group consisting of Glyl6Asp, Glu22Ala and Thr87Gln.

[0035] Embodiment 20: The vector of embodiment 19, wherein said beta globin gene comprises the mutation Glyl6Asp.

[0036] Embodiment 21 : The vector according to any one of embodiments 19-20, wherein said beta globin gene comprises the mutation Glu22Ala.

[0037] Embodiment 22: The vector according to any one of embodiments 19-21, wherein said beta globin gene comprises the mutation Thr87Gln.

[0038] Embodiment 23: The vector of embodiment 19, wherein said anti-sickling human β-globin gene comprises about 2.3 kb of recombinant human β-globin gene including exons and introns under the control of the human β-globin gene 5' promoter and the human β-globin 3 ' enhancer. [0039] Embodiment 24: The vector according to any one of embodiments 1-23, wherein said β-globin gene comprises β-globin intron 2 with a 375 bp Rsal deletion from IVS2.

[0040] Embodiment 25: The vector according to any one of embodiments 1-24, wherein said β-globin gene comprises an Sspl (S) to Rsal (R) deletion (~220bp).

[0041] Embodiment 26: The vector according to any one of embodiments 1-25, wherein said vector comprises a human Ankyrin insulator element.

[0042] Embodiment 27: The vector of embodiment 26, wherein said human

Ankyrin insulator element comprises a nucleic acid sequence having at least 90%, or at least 95%, or at least 98% sequence identity with the sequence ranging from 5325 to 5483 of SEQ ID NO:l, or from 6458 to 6616 of SEQ ID NO:2.

[0043] Embodiment 28: The vector of embodiment 27, wherein said human

Ankyrin insulator element consists of a nucleic acid sequence ranging from 5325 to 5483 of SEQ ID NO:l, or from 6458 to 6616 of SEQ ID NO:2.

[0044] Embodiment 29: The vector according to any one of embodiments 26-28, wherein said human ankyrin insulator is adjacent to HS4.

[0045] Embodiment 30: The vector according to any one of embodiments 26-28, wherein said human ankyrin insulator is adjacent to HS2.

[0046] Embodiment 31 : The vector according to any one of embodiments 1-30, wherein said vector comprises a murine GATA1-HS2.

[0047] Embodiment 32: The vector of embodiment 31, wherein said murine

GATA1-HS2 comprises a nucleic acid sequence having at least 90%, or at least 95%, or at least 98% sequence identity with the sequence ranging from 6256 to 6457 SEQ ID NO:2.

[0048] Embodiment 33: The vector of embodiment 32, wherein said murine GATA1-HS2 consists of the nucleic acid sequence ranging from 6256 to 6457 SEQ ID NO:2.

[0049] Embodiment 34: The vector according to any one of embodiments 31-33, wherein said GATA1-HS2 is adjacent to HS2.

[0050] Embodiment 35: The vector according to any one of embodiments 31-33, wherein said GATA1-HS2 is adjacent to HS4. [0051] Embodiment 36: The vector according to any one of embodiments 1-35, further comprising an insulator in the 3' LTR.

[0052] Embodiment 37: The vector of embodiment 36, wherein said insulator comprises FB (FII/BEAD-A), a 77 bp insulator element, which contains the minimal CTCF binding site enhancer-blocking components of the chicken β-globin 5' Dnasel- hypersensitive site 4 (5' HS4).

[0053] Embodiment 38: The vector according to any one of embodiments 1-37, wherein said vector comprises a ψ region vector genome packaging signal.

[0054] Embodiment 39: The vector according to any one of embodiments 1-38, wherein the 5' LTR comprises a CMV enhancer/promoter.

[0055] Embodiment 40: The vector according to any one of embodiments 1-39, wherein said vector comprises a Rev Responsive Element (RRE).

[0056] Embodiment 41 : The vector according to any one of embodiments 1-40, wherein said vector comprises a central polypurine tract.

[0057] Embodiment 42: The vector according to any one of embodiments 1-41, wherein said vector comprises a post-translational regulatory element.

[0058] Embodiment 43: The vector of embodiment 42, wherein the

posttranscriptional regulatory element is modified Woodchuck Post-transcriptional Regulatory Element (WPRE).

[0059] Embodiment 44: The vector of embodiment 1, wherein said vector comprises the nucleic acid sequence of SEQ ID NO: 1.

[0060] Embodiment 45: The vector of embodiment 1, wherein said vector comprises the nucleic acid sequence of SEQ ID NO:2.

[0061] Embodiment 46: The vector according to any one of embodiments 1-45, wherein said vector is incapable of reconstituting a wild-type lenti virus through recombination.

[0062] Embodiment 47: A recombinant lentiviral vector (LV) comprising: an expression cassette comprising a nucleic acid construct comprising:

[0063] a human β-globin locus control region, wherein at least one hypersensitive site (HS) sequence is absent; and

[0064] a recombinant human beta globin gene encoding a beta globin polypeptide; and where said LV is a TAT-independent and self-inactivating (SIN) lentiviral vector.

[0065] Embodiment 48: The vector of embodiment 47, wherein said β-globin locus control region omits hypersensitive site (HS) sequence HS4.

[0066] Embodiment 49: The vector according to any one of embodiments 47^48, wherein said β-globin locus control region comprises wildtype hypersensitive site (HS) sequence HS2.

[0067] Embodiment SO: The vector according to any one of embodiments 47-48, wherein said β-globin locus control region comprises wildtype hypersensitive site (HS) sequence HS3.

[0068] Embodiment 51: The vector according to any one of embodiments 47-48, wherein said β-globin locus control region comprises hypersensitive site (HS) core sequence HS2.

[0069] Embodiment 52: The vector according to any one of embodiments 47-48, wherein said β-globin locus control region comprises hypersensitive site (HS) core sequences HS3.

[0070] Embodiment 53: The vector according to any one of embodiments 47-48, wherein said β-globin locus control region comprises wildtype hypersensitive site (HS) sequences HS2 and HS3.

[0071] Embodiment 54: T The vector according to any one of embodiments 47-48, wherein said β-globin locus control region comprises hypersensitive site (HS) core sequences HS2 and HS3.

[0072] Embodiment 55: The vector according to any one of embodiments 47-54, wherein said HS2 core sequence consists of a nucleic acid sequence having at least 90%, or at least 95%, or at least 98% sequence identity with the sequence ranging from 5484 to 5908 of SEQ ID NO: 1 , or from 5077 to 5501 of SEQ ID NO:2.

[0073] Embodiment 56: The vector of embodiment 55, wherein said HS2 core sequence consists of a nucleic acid sequence ranging from 5484 to 5908 of SEQ ID NO:l, or from 5077 to 5501 of SEQ ID NO:2.

[0074] Embodiment 57: The vector according to any one of embodiments 47-56, wherein said HS3 core sequence consists of a nucleic acid sequence having at least 90%, or at least 95%, or at least 98% sequence identity with the sequence ranging from S909 to 6252 of SEQ ID NO: 1, or from 5502 to 5845 of SEQ ID NO:2.

[0075] Embodiment 58: The vector of embodiment 57, wherein said HS3 core sequence consists of a nucleic acid sequence ranging from 5909 to 6252 of SEQ ID NO: 1 , or from 5502 to 5845 of SEQ ID NO:2.

[0076] Embodiment 59: The vector according to any one of embodiments 47-58, wherein said human beta globin gene comprises a wild-type beta globin gene.

[0077] Embodiment 60: The vector according to any one of embodiments 47-58, wherein said human beta globin gene comprises an anti- sickling human beta globin gene encoding an anti- sickling beta globin polypeptide.

[0078] Embodiment 61 : The vector of embodiment 60, wherein said anti-sickling human beta globin gene encoding an anti-sickling-beta globin polypeptide comprise one or more mutations selected from the group consistnng of Glyl6Asp, Glu22Ala and Thr87Gln.

[0079] Embodiment 62: The vector of embodiment 61, wherein said beta globin gene comprises the mutation Glyl6Asp.

[0080] Embodiment 63: The vector according to any one of embodiments 61-62, wherein said beta globin gene comprises the mutation Glu22Ala.

[0081] Embodiment 64: The vector according to any one of embodiments 61-63, wherein said beta globin gene comprises the mutation Thr87Gln.

[0082] Embodiment 65: The vector of embodiment 61, wherein said anti-sickling human β-globin gene comprises about 2.3 kb of recombinant human β-globin gene including exons and introns under the control of the human β-globin gene 5' promoter and the human β-globin 3' enhancer.

[0083] Embodiment 66: The vector according to any one of embodiments 47-65, wherein said β -globin gene comprises β-globin intron 2 with a 375 bp Rsal deletion from IVS2.

[0084] Embodiment 67: The vector according to any one of embodiments 47-66, wherein said vector comprises a human Ankyrin insulator element.

[0085] Embodiment 68: The vector of embodiment 67, wherein said human Ankyrin insulator element comprises a nucleic acid sequence having at least 90%, or at least 95%, or at least 98% sequence identity with the sequence ranging from S32S to 5483 of SEQ ID NO:l, or from 6458 to 6616 of SEQ ID NO:2.

[0086] Embodiment 69: The vector of embodiment 68, wherein said human

Ankyrin insulator element consists of a nucleic acid sequence ranging from 5325 to 5483 of SEQ ID NO:l, or from 6458 to 6616 of SEQ ID NO:2.

[0087] Embodiment 70: The vector according to any one of embodiments 67-69, wherein said human ankyrin insulator is adjacent to HS3.

[0088] Embodiment 71 : The vector according to any one of embodiments 67-69, wherein said human ankyrin insulator is adjacent to HS2.

[0089] Embodiment 72: The vector according to any one of embodiments 47-71, wherein said vector comprises a murine GATA1-HS2.

[0090] Embodiment 73: The vector of embodiment 72, wherein said murine

GATA1-HS2 comprises a nucleic acid sequence having at least 90%, or at least 95%, or at least 98% sequence identity with the sequence ranging from 6256 to 6457 SEQ ID NO:2.

[0091] Embodiment 74: The vector of embodiment 73, wherein said murine

GATA1-HS2 consists of the nucleic acid sequence ranging from 6256 to 6457 SEQ ID NO:2.

[0092] Embodiment 75: The vector according to any one of embodiments 72-74, wherein said GATA1-HS2 is adjacent to HS2.

[0093] Embodiment 76: The vector according to any one of embodiments 72-74, wherein said GATA1-HS2 is adjacent to HS4.

[0094] Embodiment 77: The vector according to any one of embodiments 47-76, further comprising an insulator in the 3' LTR.

[0095] Embodiment 78: The vector of embodiment 77, wherein said insulator comprises FB (FII/BEAD-A), a 77 bp insulator element, which contains the minimal CTCF binding site enhancer-blocking components of the chicken β-globin 5' Dnasel- hypersensitive site 4 (5' HS4).

[0096] Embodiment 79: The vector according to any one of embodiments 47-78, wherein said vector comprises a ψ region vector genome packaging signal.

[0097] Embodiment 80: The vector according to any one of embodiments 47-79, wherein the 5' LTR comprises a CMV enhancer/promoter. [0098] Embodiment 81: The vector according to any one of embodiments 47-80, wherein said vector comprises a Rev Responsive Element (RRE).

[0099] Embodiment 82: The vector according to any one of embodiments 47-81, wherein said vector comprises a central polypurine tract.

[0100] Embodiment 83: The vector according to any one of embodiments 47-82, wherein said vector comprises a post-translational regulatory element.

[0101] Embodiment 84: The vector of embodiment 83, wherein the

posttranscriptional regulatory element is modified Woodchuck Post-transcriptional Regulatory Element (WPRE).

[0102] Embodiment 85: The vector according to any one of embodiments 47-84, wherein said vector is incapable of reconstituting a wild-type lenti virus through

recombination.

[0103] Embodiment 86: A host cell transduced with a vector according to any one of embodiments 1-85.

[0104] Embodiment 87: The host cell of embodiment 86, wherein the cell is a stem cell.

[0105] Embodiment 88: The host cell of embodiment 87, wherein said cell is a stem cell derived from bone marrow, or from monilized peripheral blood, or from umbilical blood. In certain embodiments the stem cell is not an embryonic stem cell.

[0106] Embodiment 89: The host cell of embodiment 86, wherein the cell is a 293T cell.

[0107] Embodiment 90: The host cell of embodiment 86, wherein, wherein the cell is a human hematopoietic progenitor cell.

[0108] Embodiment 91 : The host cell of embodiment 90, wherein the human hematopoietic progenitor cell is a CD34+ cell.

[0109] Embodiment 92: A method of treating a hemoglobinopathy, in a subject, said method comprising:

[0110] transducing a stem cell and/or progenitor cell from said subject with a vector according to any one of embodiments 1-85; and

[0111 ] transplanting said transduced cell or cells derived therefrom into said subject where said cells or derivatives therefrom express said anti-sickling human beta globin gene.

[0112] Embodiment 93: The method of embodiment 92, wherein the cell is a stem cell.

[0113] Embodiment 94: The host cell of embodiment 92, wherein said cell is a stem cell derived from bone marrow.

[0114] Embodiment 95 : The method of embodiment 92, wherein, wherein the cell is a human hematopoietic progenitor cell.

[0115] Embodiment 96: The method of embodiment 95, wherein the human hematopoietic progenitor cell is a CD34 + cell.

[0116] Embodiment 97: The method according to any one of embodiments 92-96, wherein said hemoglobinopathy is sickle cell disease.

[0117] Embodiment 98: The method according to any one of embodiments 92-96, wherein said hemoglobinopathy is β-thalassemia. DEFINITIONS

[0118] An "HS core sequence" as used herein refers to a reduced β-globin locous control region (LCR) hypersensitivity site (HS) sequence as defined herein (e.g., (HS2 (-420 bp), HS3 (~340bp), and/or HS4 (-410 bp)). Full-length HS sequences refers to LCR HS2, HS3, and HS4 as previously defined (e.g., HS2 (~1.20kb), HS3 (~1.28kb), and HS4 (-l.lkb)) (see, e.g., Forrester et al. (1986) Proc. Natl Acad. Sci. USA, 83: 1359-1363).

[0119] "Recombinant" is used consistently with its usage in the art to refer to a nucleic acid sequence that comprises portions that do not naturally occur together as part of a single sequence or that have been rearranged relative to a naturally occurring sequence. A recombinant nucleic acid is created by a process that involves the hand of man and/or is generated from a nucleic acid that was created by hand of man (e.g. , by one or more cycles of replication, amplification, transcription, etc.). A recombinant virus is one that comprises a recombinant nucleic acid. A recombinant cell is one that comprises a recombinant nucleic acid.

[0120] As used herein, the term "recombinant lenti viral vector" or "recombinant LV) refers to an artificially created polynucleotide vector assembled from an LV and a plurality of additional segments as a result of human intervention and manipulation. [0121] By "globin nucleic acid molecule" is meant a nucleic acid molecule that encodes a globin polypeptide. In various embodiments the globin nucleic acid molecule may include regulatory sequences upstream and/or downstream of the coding sequence.

[0122] By "globin polypeptide" is meant a protein having at least 85%, or at least 90%, or at least 95%, or at least 98% amino acid sequence identity to a human alpha, beta or gamma globin.

[0123] The term "therapeutic functional globin gene" refers to a nucleotide sequence the expression of which leads to a globin that does not produce a hemoglobinopathy phenotype, and which is effective to provide therapeutic benefits to an individual with a defective globin gene. The functional globin gene may encode a wild-type globin appropriate for a mammalian individual to be treated, or it may be a mutant form of globin, preferably one which provides for superior properties, for example superior oxygen transport properties or anti- sickling properties. The functional globin gene includes both exons and introns, as well as globin promoters and splice donors/acceptors.

[0124] By "an effective amount" is meant the amount of a required agent or composition comprising the agent to ameliorate or eliminate symptoms of a disease relative to an untreated patient. The effective amount of composition^) used to practice the methods described herein for therapeutic treatment of a disease varies depending upon the manner of administration, the age, body weight, and general health of the subject.

Ultimately, the attending physician or veterinarian will decide the appropriate amount and dosage regimen. Such amount is referred to as an "effective" amount.

[0125] The term "sequence identity" refers to the extent to which two optimally aligned polynucleotide or polypeptide sequences are invariant throughout a window of alignment of components, e.g., nucleotides or amino acids. "Identity" can be readily calculated by known methods including, but not limited to, those described in:

Computational Molecular Biology (Lesk, A. M., ed.) Oxford University Press, New York (1988); Biocomputing: Informatics and Genome Projects (Smith, D. W., ed.) Academic Press, New York (1993); Computer Analysis of Sequence Data, Part I (Griffin, A. M., and Griffin, H. G., eds.) Humana Press, New Jersey (1994); Sequence Analysis in Molecular Biology (von Heinje, G., ed.) Academic Press (1987); and Sequence Analysis Primer (Gribskov, M. and Devereux, J., eds.) Stockton Press, New York (1991); and the like.

[0126] An "identity fraction" for aligned segments of a test sequence and a reference sequence is the number of identical components which are shared by the two aligned sequences divided by the total number of components in the reference sequence segment, i.e., the entire reference sequence or a smaller defined part of the reference sequence.

Percent sequence identity is typically represented as the identity fraction multiplied by 100. As used herein, the term "percent sequence identity" or "percent identity" refers to the percentage of identical nucleotides in a linear polynucleotide sequence of a reference ("query") polynucleotide molecule (or its complementary strand) as compared to a test ("subject") polynucleotide molecule (or its complementary strand) when the two sequences are optimally aligned (with appropriate nucleotide insertions, deletions, or gaps totaling less than 20 percent of the reference sequence over the window of comparison).

[0127] Optimal alignment of sequences for aligning a comparison window is well known to those skilled in the art and may be conducted by tools such as the local homology algorithm of Smith and Waterman, the homology alignment algorithm of Needleman and Wunsch, the search for similarity method of Pearson and Lipman, and optionally by computerized implementations of these algorithms such as GAP, BESTFIT, FAST A, and TFASTA available as part of the GCG® Wisconsin Package® (Accelrys Inc., Burlington, Mass.). The comparison of one or more polynucleotide sequences may be to a full-length polynucleotide sequence or a portion thereof, or to a longer polynucleotide sequence. In various embodiments "percent identity" may also be determined using BLASTX version 2.0 for translated nucleotide sequences and BLASTN version 2.0 for polynucleotide sequences.

[0128] The percent of sequence identity can be determined using the "Best Fit" or

"Gap" program of the Sequence Analysis Software Package. TM. (Version 10; Genetics Computer Group, Inc., Madison, Wis.). "Gap" utilizes the algorithm of Needleman and Wunsch (1970) J. Mol. Biol 48: 443-453, to find the alignment of two sequences that maximizes the number of matches and minimizes the number of gaps. "BestFit" performs an optimal alignment of the best segment of similarity between two sequences and inserts gaps to maximize the number of matches using the local homology algorithm of Smith and Waterman (1981) Adv. Appl. Math., 2: 482-489, Smith et al. (1983) Nucleic Acids Res. 11 : 2205-2220. Useful methods for determining sequence identity are also disclosed in Guide to Huge Computers (Martin J. Bishop, ed., Academic Press, San Diego (1994)), and Carillo et al. (1988) Appl. Math, 48: 1073). Certain illustrative computer programs for detennining sequence identity include, but are not limited to, the Basic Local Alignment Search Tool (BLAST) programs, that are publicly available from National Center Biotechnology Information (NCBI) at the National Library of Medicine, National Institute of Health, Bethesda, Md. 20894; see BLAST Manual, Altschul et al., NCBI, NLM, NIH; (Altschul et al, J. Mol. Biol. 215:403-410 (1990)); version 2.0 or higher of BLAST programs allows the introduction of gaps (deletions and insertions) into alignments; for peptide sequence, BLASTX can be used to determine sequence identity; and for polynucleotide sequence, BLASTN can be used to determine sequence identity. BRIEF DESCRIPTION OF THE DRAWINGS

[0129] Figure 1 A schematically illustrates a comparison of the structures of CCL- βAS3-FB with CCL-GLOBE1-AS3. Figure IB shows a comparison of the two vectors with respect to raw titer (top) and transduction in erythroid (middle) and myeloid (bottom) cell lines. VCN=vector copy number

[0130] Figure 2 schematically illustrates various strategies investigated in designing a series of CCLc-βAS3-FB derivatives.

[0131] Figure 3A shows the average fold difference in titers for various ΔHS4 constructs (CCLc-βAS3-FB , CCLc-βAS3-FB-(HS4), and CCLc-βAS3-FB-(- HS4)(+HPRT)), while Figure 3B shows the effect of vector length on gene transfer and Figure 3C shows the expression levels of the various constructs.

[0132] Figure 4 schematically illustrates the structure of CCLc-βAS3-FB (8.9kb)

(top), CCLc-βAS3-FB-(ΔHS4) (6.3 kb) (middle), and CCL-GLOBE1-AS3 (6.3 kb) (bottom).

[0133] Figure 5A-5C illustrate the titers of CCLc-Globel-FB, CCLc-βAS3-FB- (ΔHS4), and CCLC-βAS3-FB (Fig. 5A), the transduction levels of these vectors (Fig. 5B), and expression levels (Fig. SC).

[0134] Figure 6A shows the raw titers plotted as a function of construct size for IS different lenti viral constructs. Figure 6B shows the expression levels (vector copy number (VCN) plotted as a functions of construct size for these different lenti viral constructs

[0135] Figure 7 illustrates "open chromatin" track sets, that combine DNasel hypersensitivity, formaldehyde-assisted isolation of regulatory elements, and chromatin immunoprecipitation data to identify accessible chromatin regions, that was combined with the "transcription Factor ChlP-seq" and "histone modification" track sets used to generate correct boundaries for the LCR's HS core sequences.

[0136] Figures 8A and 8B schematically illustrates novel defined LCR HS core sequences (HS2(~420 bp), HS3(~340bp), HS4(~410 bp)) that were then used to replace the putative LCR HS sequences present within the "mini-LCR" (~3.6 kb reduced to ~1.2 kb) to produce an "optimized mini-LCR" (CoieLCR-Opti).

[0137] Figure 9 illustrates the relationship between raw titer and construct size.

[0138] Figure 10 schematically illustrates CCLc-p AS3-(mGATA-HS2/ANK)- (ΔIVS2)-(CoreLCR)-(Opti.)-FB and CCLc-βAS3-(+Ank)-(CoreLCR)-(Opti.)-FB vectors.

[0139] Figure 11 shows raw titer produced by CCLc-βAS3-FB and CCLc-βAS3-

(+Ank)-CoreLCR(Opti)-FB constructs.

[0140] Figure 12 (top panel) shows average fold difference in raw titer produced by

CCLc-βAS3-FB andCCLc-βAS3-(+Ank)-CoreLCR(Opti)-FB constructs. Figure 12 (bottom panel) shows average % mRNA/VCN produced using CCLc-βAS3-FB and CCLc- βAS3-(+Ank)-CoreLCR(Opti)-FB constructs.

[0141] Figure 13 shows the effect of replacing LCR with ANK and HS2, HS3, HS4 optimized core elements.

[0142] Figure 14 shows the effect of replacing LCR with ANK and HS2, HS3, HS4 core elements.

[0143] Figure 15 A schematically illustrates the pCCLC-p AS3-FB(+CoreLCR w-

Ank-R) optimized vector ((10528 bp). Figure 15B illustrates the nucleic acid sequence of the vector (SEQ ID NO: 1).

[0144] Figure 16A illustrates the pCCL-βAS3-(4tnGATA-HS2-Ank-dIVS2- CoreLCRopti)-FB optimized vector (10483 bp). Figure 16B illustrates the nucleic acid sequence of the vector (SEQ ID NO:2).

[0145] Figure 17 schematically illustrates novel defined LCR HS core sequences

(HS2 (-420 bp), HS3(~340bp), and HS4 (-410 bp)) that were used to replace the putative LCR HS sequences present within the "mini-LCR" (~3.6 Kb reduced to ~1.2 Kb) to produce an "optimized mini-LCR".

[0146] Figures 18 A- 18C, illustrate various constructs adding element to the

"optimized mini-LCR". Fig. 18A) CCLc-βAS3-FB-mGATA/ANK-CoreLCR Opti- AHS234/ΔIVS2. Note MVS2 references an Sspl (S) to Rsal (R) deletion (~220bp), e.g., as described by Antoniou et al. (1998) Nucl Acids Res., 26(3): 721-729. Fig. 18B) CC1C- βAS3-FB-CorLCR Opti-ANK. Fig. 18C) mGATAl-HS2 and the human ankyrin insulator elements in alternative configurations in combination with the "optimized mini-LCR (e.g., CCLc-βAS3-FB-CoreLCR Opti-mGATA/ANK).

[0147] Figure 19 schematically illustrates mGATAl-HS2 and human ankyrin insulator elemets used in a construct with full-length erythroid enhancers (HS2 and HS3).

[0148] Figure 20 illustrates head-to-head raw titer produced using different constructs.

[0149] Figure 21 illustrates the results of gene transfer studies. Escalating viral dose

- CD34+ BM HSPCs cultured under myeloid conditions.

[0150] Figure 22 illustrates the results of erythroid expression studies. Constant viral dose of 2x 10 7 (TU/ml) - CD34+ BM HSPCs cultured under erythroid conditions.

[0151] Figure 23 illustrates the results of gene transfer studies to determine effect of placing mGATA/ANK in alternative configurations. Constant viral dose of 1 x 10 7 (TU/ml)

- CD34+ BM HSPCs cultured under myeloid conditions.

[0152] Figure 24 illustrates the results of expression studies to determine effect of placing mGATA/ANK in alternative configurations. Constant viral dose of 6.6 x 10 6 or 1 x 10 7 (TU/ml) - CD34+ BM HSPCs cultured under erythroid conditions.

DETAILED DESCRIPTION

[0153] Sickle cell disease (SCD) is a multisystem disease, associated with severe episodes of acute illness and progressive organ damage, and is one of the most common monogenic disorders worldwide. Because SCD results from abnormalities in rbc, which in turn are produced from adult HSC, HSCT from a healthy (allogeneic) donor can benefit patients with SCD, by providing a source for life-long production of normal red blood cells. However, allogeneic HSCT is limited by the availability of well-matched donors and by immunological complications of graft rejection and graft- versus-host disease.

[0154] It is believed that autologous stem cell gene therapy for SCD has the potential to treat this illness without the need for immune suppression of current allogeneic HSCT approaches. In particular, it is believed that autologous stem cell gene therapy that introduces anti- sickling human beta globin into hematopoietic cells (or progenitors thereof) can provide effective therapy for SCD (including, for example, normalized rbc physiology and prevention of the manifestations of SCD). [0155] Current B-globin expression vectors, however, suffer from low vector titer and sub-optimal gene transfer to hematopoietic stem cells, representing a major barrier toward the effective implementation of this gene therapy strategy to the clinic. In view of the data presented herein, it is believed that the predominant factor most likely affecting vector performance is overall vector length. We found that reduction of the "mini-LCR" by ~66% through redefining the LCR HS core sequences and, in certain embodiments, inclusion of the GATA1-H52 and/or Ankyrin insulator elements allowed for approximately 3-fold higher titer, superior gene transfer to hematopoietic stem cells and comparable expression of BAS3 when compared to the antecedent vector.

[0156] Accordingly, in various embodiments, an improved LV is provided for the introduction of a normal wild-type or an anti-sickling beta globin into stem and progenitor cells (e.g., hematopoietic stem and progenitor cells) that can then be transplanted into a subject in need thereof (e.g., a subject that has the sickle cell mutation).

[0157] We have engineered an optimized derivative of CCLc-βAS3-FB termed (CCLc-mGata/ANK-CoreLCR-β AS3-FB), that is capable of driving lineage-restricted expression of an anti-sickling B-globin like gene (βAS3). β-globin expression vectors generally contain fragments of the human β-globin Locus Control Region (LCR). The native LCR is a -20 kilo base (kb) element that regulates the temporal and erythroid- specific expression of the globin gene cluster located nearly 30-60 kb upstream from the B- globin gene promoter sequences. Investigations conducted in the 1990's elucidated the function of the native LCR through examining the effect of sequence deletion on developmental eiythropoiesis in murine mouse models. These mouse knockout studies utilized a limited repertoire of restriction enzymes to define and delete those sequences found to be essential for LCR function. It was later discovered that inclusion of these roughly defined LCR sequences, specifically hypersensitive sites (HS) HS2, HS3 and/or HS4, were essential to sustained, high-level and erythroid- specific expression of β-globin in the lentiviral vector system. Grouping the LCR's roughly defined HS2 (1 .20kb), HS3 (1.28kb), and HS4 (l.lkb) sequences together to form a "mini-LCR", faithfully conferred lineage-restricted expression of BAS3 to CCLc~βAS3-FB when placed upstream of a minimal promoter. However, the large size of the "mini-LCR" has a detrimental effect on packaging efficiency and gene transfer to human hematopoietic stem cells (HSC), limiting the effective deployment of this lentiviral vector to the clinic for gene therapy of hemoglobinopathies. [0158] Optimization of the "mini-LCR" (with the primary goal of reducing length) was accomplished through redefining the putative boundaries of the LCR's HS core sequences using published genomic data available through ENCODE (Accessible via the UCSC Genome Browser). For example, the "Open Chromatin" track sets, which combine DNasel hypersensitivity, Formaldehyde-Assisted Isolation of Regulatory Elements, and chromatin irnmunoprecipitation data to identify accessible chromatin regions, was combined with the "transcription Factor ChlP-seq" and "histone modification" track sets to generate new boundaries for the LCR's HS core sequences. These novel defined LCR HS core sequences (HS2(~420 bp), HS3(~340bp), HS4(~410 bp)) were then used to replace the putative LCR HS sequences present within the "mini-LCR" (~3.6 kb reduced to ~1.1 kb) to produce an "optimized mini-LCR". In addition, we added elements to the "optimized mini- LCR" known to facilitate position independent expression of β-globin such as the murine GATA1-HS2 (~220 bp) and/or the human Ankyrin insulator (-150 bp) elements. These vectors, rationally-designed for reduced sizes of the LCR fragments and added

transcriptional enhancing elements are believed to be produced at higher titers than the original β-globin lentiviral vector and have improved gene transfer to human HSC while retaining strong erythroid-specific gene expression. Such improved lentiviral vectors can be effective for gene therapy of hemoglobinopathies such as sickle cell disease and β- thalassemia.

[0159] In certain embodiments the lentiviral vectors (LVs) comprise an expression cassette comprising a nucleic acid construct comprising a human β-globin locus control region comprising at least one hypersensitive site (HS) core sequence selected from the group consisting of HS2 (~420bp), HS3 (~ 340 bp), and HS4 (~410bp), and a recombinant human beta globin gene encoding a beta globin polypeptide. Typically, the lentiviral vector is a TAT-independent and self-inactivating (SUN) lentiviral vector.

[0160] In certain embodiments the β-globin locus control region comprises hypersensitive site (HS) core sequence HS2. In certain embodiments the β-globin locus control region comprises hypersensitive site (HS) core sequence HS3. In certain embodiments the β-globin locus control region comprises hypersensitive site (HS) core sequence HS4. In certain embodiments the β-globin locus control region comprises hypersensitive site (HS) core sequences HS2 and HS3. In certain embodiments the β-globin locus control region comprises hypersensitive site (HS) core sequences HS2 and HS4. In certain embodiments the β-globin locus control region comprises hypersensitive site (HS) core sequences HS3 and HS4. In certain embodiments the β-globin locus control region comprises hypersensitive site (HS) core sequences HS2, HS3 and HS4.

[0161] In certain embodiments the HS2 core sequence consists of a nucleic acid sequence having having at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 98% sequence identity with the sequence ranging from 5484 to 5908 of SEQ ID NO: 1 , or from 5077 to 5501 of SEQ ID NO: 2. In certain embodiments the HS2 core sequence consists of a nucleic acid sequence ranging from 5484 to 5908 of SEQ ID NO: 1, or from 5077 to 5501 of SEQ ID NO: 2.

[0162] In certain embodiments the HS3 core sequence consists of a nucleic acid sequence having at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 98% sequence identity with the sequence ranging from 5909 to 6252 of SEQ ID NO: 1, or from 5502 to 5845 of SEQ ID NO:2. In certain embodiments the HS3 core sequence consists of a nucleic acid sequence ranging from 5909 to 6252 of SEQ ID NO: 1, or from 5502 to 5845 of SEQ ID NO: 2.

[0163] In certain embodiments the HS4 core sequence consists of a nucleic acid sequence having at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 98% sequence identity with the sequence ranging from 6253 to 6662 of SEQ ID NO: 1, or from 5846 to 6255 of SEQ ID NO:2. In certain embodiments the HS4 core sequence consists of a nucleic acid sequence ranging from 6253 to 6662 of SEQ ID NO:l, or from 5846 to 6255 ofSEQ IDNO: 2.

[0164] In certain embodiments the β-globin locus control region comprises the nucleic acid sequence ranging from 5484 to 6662 of SEQ ID NO:l, or from 5077 to 6255 of SEQ ID NO: 2. In certain embodiments the human β-globin locus control region consists of the nucleic acid sequence ranging from 5484 to 6662 of SEQ ID NO: 1, or from 5077 to 6255 ofSEQ ID NO:2.

[0165] In certain embodiments, one or more of the HS core sequences can be used in combination with a full-length {e.g., wildtype) HS sequence. Thus, for example, in certain embodiments, a core HS2 may be used in combination with a core HS3, or with a wildtype HS3, or with a core HS4 or a wildtype HS4, or with a core HS3 and a wildtype HS4, or with a wildtype HS3 and a core HS4. In certain embodiments, a core HS3 may be used in combination with a core HS2, or with a wildtype HS2, or with a core HS4 or a wildtype HS4, or with a core HS2 and a wildtype HS4, or with a wildtype HS2 and a core HS4. Thus, for example, in certain embodiments, a core HS4 may be used in combination with a core HS2, or with a wildtype HS2, or with a core HS3 or a wildtype HS3, or with a core HS2 and a wildtype HS3, or with a wildtype HS2 and a core HS3. In certain embodiments an HS4 is absent Accordingly, the present hypersensitive sites can be a core HS2, and a core HS3, or a wildtype HS2 and a core HS3, or a core HS2 and a wildtype HS3. In certain embodiments, particularly where a mGATAlq-HS2 and/or human ankyrin insulator element is present, the hypersensitive sites can comprise or consist of a wildtype HS3 and a wildtype HS2.

[0166] In certain embodiments the human beta globin gene in the vectors contemplated herein comprises an anti-sickling human beta globin gene encoding an anti- sickling beta globin polypeptide. In certain embodiments the anti-sickling version of a human beta globin gene used in the vector comprises one, two, or mree mutations selected from the group consisting of Glyl6Asp, Glu22Ala and Thr87Gln (see, e.g., Levasseur (2004) J. Biol Chem. 279(26): 27518-27524). Without being bound to a particular theory, it is believed the Glu22Ala mutation increases affinity to a-chain, the Thr87Gln mutation blocks lateral contact with Val6 of βS protein, and the Gly 16Asp mutation decreases axial contact between globin chains.

[0167] In certain embodiments the vectors described herein comprise a human

Ankyrin insulator element. In certain embodiments the Ankyrin insulator element comprises a nucleic acid sequence having at least 90%, or at least 95%, or at least 98% sequence identity with the sequence ranging from 5325 to 5483 of SEQ ID NO: 1 , or from 6458 to 6616 of SEQ ID NO:2. In certain embodiments the human Ankyrin insulator element consists of a nucleic acid sequence ranging from 5325 to 5483 of SEQ ID NO:l, or from 6458 to 6616 of SEQ ID NO:2.

[0168] In certain embodiments the vectors described herein comprise a murine GATA1-HS2. In certain embodiments the murine GATA1-HS2 (mGATA-HS2) comprises a nucleic acid sequence having at least 90%, or at least 95%, or at least 98% sequence identity with the sequence ranging from 6256 to 6457 SEQ ID NO:2. In certain

embodiments the murine GATA1-HS2 consists of the nucleic acid sequence ranging from 6256 to 6457 SEQ ID NO: 2.

[0169] In various embodiments, the LVs described herein can have additional safety features not included in previous β-globin encoding lentiviral constructs. In certain embodiments, these features include the presence of an insulator (eg., an FB insulator in the 3'LTR). Additionally, or alternatively, in certain embodiments, the HIV LTR has been substituted with an alternative promoter {e.g., a CMV) to yield a higher titer vector without the inclusion of the HIV TAT protein during packaging. Other strong promoters (e.g., RSV, and the like can also be used).

[0170] In certain embodiments the vectors contemplated herein include the various elements shown in the vector illustrated in Figure 15 A. In certain embodiments the vectors contemplated herein comprise the nucleic acid shown in Figure 15B (SEQ ID NO:l).

[0171] In certain embodiments the vectors contemplated herein include the various elements shown in the vector illustrated in Figure 16 A. In certain embodiments the vectors contemplated herein comprise the nucleic acid shown in Figure 16B (SEQ ID NO:2).

include the various elements shown in the vector illustrated in Figure ISA.

[0172] As shown in the Examples provided herein the vectors described herein are effective to transduce cells at high titer and to also provide high levels of expression.

[0173] In view of these results, it is believed that LVs described herein, e.g. , recombinant TAT-independent, SIN LVs that express a human beta-globin gene can be used to effectively treat hemoglobinopathies in subjects (eg., human and non-human mammals). Such hemoglobinopathies include, but are not limited to sickle cell disease (SCD) and β- thalassemia. It is believed these vectors can be used for the modification of stem cells (e.g., hematopoietic stem and progenitor cells) that can be introduced into a subject in need thereof for the treatment of, e.g., SCD or β-thalassemia. Moreover, it appears that the resulting cells will produce enough of the transgenic β-globin protein to demonstrate significant improvement in subject health. It is also believed the vectors can be directly administered to a subject to achieve in vivo transduction of the target (e.g., hematopoietic stem or progenitor cells) and thereby also effect a treatment of subjects in need thereof.

[0174] As noted above, in various embodiments the LVs described herein can comprise various safety features. For example, the HIV LTR has been substituted with a CMV promoter to yield higher titer vector without the inclusion of the HIV TAT protein during packaging. In certain embodiments an insulator (e.g., the FB insulator) is introduced into the 3 "LTR for safety. The LVs are also constructed to provide efficient transduction and high titer.

[0175] It will be appreciated that the foregoing elements are illustrative and need not be limiting. In view of the teachings provided herein, suitable substitutions for these elements will be recognized by one of skill in the art and are contemplated within the scope of the teachings provided herein. Anti-sickling beta globin gene and expression cassette,

[0176] As indicated above, in various embodiments the LV described herein comprise an expression cassette encoding a wild-type β-globin gene, or an anti-sickling human β-globin gene. On illustrative, but non-limiting cassette is βAS3 which comprises an -2.3 kb recombinant human β-globin gene (exons and introns) with three amino acid substitutions (Thr87Gln; Glyl6Asp; and Glu22Ala) under the control of transcriptional control elements (e.g., the human β-globin gene 5' promoter (e.g., ~266 bp), the human β- globin 3' enhancer (e.g., -260 bp ), β-globin intron 2 with a -375 bp Rsal deletion from IVS2, and a -3.4 kb composite human β-globin locus control region (e.g., HS2 -1203 bp; HS3 -1213 bp; HS4 -954 bp). One embodiment of a βAS3 cassette is described by Levasseur (2003) Blood 102: 4312-4319.

[0177] In certain embodiments the β-globin gene comprises an an Sspl (S) to Rsal

(R) deletion (~220bp), e.g., as described by Antoniou et al. 1998) Nucl. Acids Res., 26(3): 721-729 (see, e.g., pCCL-BAS3-FB(+CORE ΔIVS2) optimized, SEQ ID NO:3).

[0178] The βAS3 cassette, however, is illustrative and need not be limiting. Using the known cassette described herein (see, e.g., Example 1), numerous variations will be available to one of skill in the art Such variations include, for example, use of a gene encoding a wild-type β-globin, use of a gene comprising one or two mutations selected from the group consisting of Thr87Gln, Glyl6Asp, and Glu22Ala, and/or further or alternative mutations to the β-globin to further enhance non- sickling properties, alterations in the transcriptional control elements (e.g., promoter and/or enhancer), variations on the intron size/structure, and the like.

TAT-Indeoendent and Self inactivating lentiviral vectors.

[0179] To further improve safety, in various embodiments, the LVs described herein comprise a TAT-independent, self-inactivating (SIN) configuration. Thus, in various embodiments it is desirable to employ in the LVs described herein an LTR region that has reduced promoter activity relative to wild-type LTR. Such constructs can be provided that are effectively ''self-inactivating" (SIN) which provides a biosafety feature. SIN vectors are ones in which the production of full-length vector RNA in transduced cells is greatly reduced or abolished altogether. This feature minimizes the risk that replication-competent recombinants (RCRs) will emerge. Furthermore, it reduces the risk that that cellular coding sequences located adjacent to the vector integration site will be aberrantly expressed. [0180] Furthermore, a SIN design reduces the possibility of interference between the

LTR and the promoter that is driving the expression of the transgene. SIN LVs can often permit Ml activity of the internal promoter.

[0181] The SIN design increases the biosafety of the LVs. The majority of the HIV LTR is comprised of the U3 sequences. The U3 region contains the enhancer and promoter elements that modulate basal and induced expression of the HIV genome in infected cells and in response to cell activation. Several of these promoter elements are essential for viral replication. Some of the enhancer elements are highly conserved among viral isolates and have been implicated as critical virulence factors in viral pathogenesis. The enhancer elements may act to influence replication rates in the different cellular target of the virus

[0182] As viral transcription starts at the 3' end of the U3 region of the 5' LTR, those sequences are not part of the viral mRNA and a copy thereof from the 3' LTR acts as template for the generation of both LTR's in the integrated provims. If the 3' copy of the U3 region is altered in a retroviral vector construct, the vector RNA is still produced from the intact 5' LTR in producer cells, but cannot be regenerated in target cells. Transduction of such a vector results in the inactivation of both LTR's in the progeny virus. Thus, the retrovirus is self-inactivating (SUN) and those vectors are known as SIN transfer vectors.

[0183] In certain embodiments self-inactivation is achieved through the introduction of a deletion in the U3 region of the 3' LTR of the vector DNA, i.e., the DNA used to produce the vector RNA. During RT, this deletion is transferred to the 5' LTR of the proviral DNA. Typically, it is desirable to eliminate enough of the U3 sequence to greatly diminish or abolish altogether the transcriptional activity of the LTR, thereby greatly diminishing or abolishing the production of full-length vector RNA in transduced cells. However, it is generally desirable to retain those elements of the LTR that are involved in polyadenylation of the viral RNA, a function typically spread out over U3, R and U5.

Accordingly, in certain embodiments, it is desirable to eliminate as many of the

transcriptionally important motifs from the LTR as possible while sparing the

polyadenylation determinants.

[0184] The SIN design is described in detail in Zufferey et al (1998) J Virol 72(12): 9873-9880, and in U.S. Patent No: 5,994,136. As described therein, there are, however, limits to the extent of the deletion at the 3' LTR. First, the 5' end of the U3 region serves another essential function in vector transfer, being required for integration (terminal dinucleotide+att sequence). Thus, the terminal dinucleotide and the att sequence may represent the 5' boundary of the U3 sequences which can be deleted. In addition, some loosely defined regions may influence the activity of the downstream polyadenylation site in the R region. Excessive deletion of U3 sequence from the 3 "LTR may decrease polyadenylation of vector transcripts with adverse consequences both on the titer of the vector in producer cells and the transgene expression in target cells.

[0185] Additional SIN designs are described in U.S. Patent Publication No:

2003/0039636. As described therein, in certain embodiments, the lentiviral sequences removed from the LTRs are replaced with comparable sequences from a non-lentiviral retrovirus, thereby forming hybrid LTRs. In particular, the lentiviral R region within the LTR can be replaced in whole or in part by the R region from a non-lentiviral retrovirus. In certain embodiments, the lentiviral TAR sequence, a sequence which interacts with TAT protein to enhance viral replication, is removed, preferably in whole, from the R region. The TAR sequence is then replaced with a comparable portion of the R region from a non- lentiviral retrovirus, thereby forming a hybrid R region. The LTRs can be further modified to remove and/or replace with non-lentiviral sequences all or a portion of the lentiviral U3 and US regions.

[0186] Accordingly, in certain embodiments, the SIN configuration provides a retroviral LTR comprising a hybrid lentiviral R region that lacks all or a portion of its TAR sequence, thereby eliminating any possible activation by TAT, wherein the TAR sequence or portion thereof is replaced by a comparable portion of the R region from a non-lentiviral retrovirus, thereby forming a hybrid R region. In a particular embodiment, the retroviral LTR comprises a hybrid R region, wherein the hybrid R region comprises a portion of the HIV R region (e.g., a portion comprising or consisting of the nucleotide sequence shown in SEQ ID NO: 10 in US 2003/0039636) lacking the TAR sequence, and a portion of the MoMSV R region {e.g., a portion comprising or consisting of the nucleotide sequence shown in SEQ ID NO: 9 in 2003/0039636) comparable to the TAR sequence lacking from the HIV R region. In another particular embodiment, the entire hybrid R region comprises or consists of the nucleotide sequence shown in SEQ ID NO: 11 in 2003/0039636.

[0187] Suitable lentiviruses from which the R region can be derived include, for example, HIV (HlV-1 and HIV-2), EIV, SIV and FIV. Suitable retroviruses from which non-lentiviral sequences can be derived include, for example, MoMSV, MoMLV, Friend, MSCV, RSV and Spumaviruses. In one illustrative embodiment, the lenti virus is HIV and the non-lentiviral retrovirus is MoMSV. [0188] In another embodiment described in US 2003/0039636, the LTR comprising a hybrid R region is a left (5*) LTR and further comprises a promoter sequence upstream from the hybrid R region. Preferred promoters are non-lenti viral in origin and include, for example, the U3 region from a non-lentiviral retrovirus {e.g., the MoMSV U3 region). In one particular embodiment, the U3 region comprises the nucleotide sequence shown in SEQ ID NO: 12 in US 2003/0039636. In another embodiment, the left (5*) LTR further comprises a lenti viral US region downstream from the hybrid R region. In one

embodiment, the US region is the HIV US region including the HIV att site necessary for genomic integration. In another embodiment, the US region comprises the nucleotide sequence shown in SEQ ID NO: 13 in US 2003/0039636. In yet another embodiment, the entire left (5') hybrid LTR comprises the nucleotide sequence shown in SEQ ID NO: 1 in US 2003/0039636.

[0189] In another illustrative embodiment, the LTR comprising a hybrid R region is a right (3*) LTR and further comprises a modified (e.g., truncated) lentiviral U3 region upstream from the hybrid R region. The modified lentiviral U3 region can include the att sequence, but lack any sequences having promoter activity, thereby causing the vector to be SIN in that viral transcription cannot go beyond the first round of replication following chromosomal integration. In a particular embodiment, the modified lentiviral U3 region upstream from the hybrid R region consists of the 3' end of a lentiviral {e.g. , HTV) U3 region up to and including the lentiviral U3 att site. In one embodiment, the U3 region comprises the nucleotide sequence shown in SEQ ID NO: IS in US 2003/0039636. In another embodiment, the right (3') LTR further comprises a polyadenylation sequence downstream from the hybrid R region. In another embodiment, the polyadenylation sequence comprises the nucleotide sequence shown in SEQ ID NO: 16 in US

2003/0039636. In yet another embodiment, the entire right (5) LTR comprises the nucleotide sequence shown in SEQ ID NO: 2 or 17 of US 2003/0039636.

[0190] Thus, in the case of HIV based LV, it has been discovered that such vectors tolerate significant U3 deletions, including the removal of the LTR TATA box {e.g., deletions from -418 to -18), without significant reductions in vector titers. These deletions render the LTR region substantially transcriptionally inactive in that the transcriptional ability of the LTR in reduced to about 90% or lower.

[0191] It has also been demonstrated that the trans-acting function of Tat becomes dispensable if part of the upstream LTR in the transfer vector construct is replaced by constitutively active promoter sequences (see, e.g., Dull et al (1998) J Virol 72(11): 8463- 8471. Furthermore, we show that the expression of rev in trans allows the production of high-titer HIV-derived vector stocks from a packaging construct which contains only gag and pol. This design makes the expression of the packaging functions conditional on complementation available only in producer cells. The resulting gene delivery system, conserves only three of the nine genes of HIV- 1 and relies on four separate transcriptional units for the production of transducing particles.

[0192] In one embodiments illustrated in Example 1, the cassette expressing an anti- sickling β-globin {e.g., βAS3) is placed in the pCCL LV backbone, which is a SIN vector with the CMV enhancer/promoter substituted in the 5' LTR.

[0193] It will be recognized that the CMV promoter typically provides a high level of non-tissue specific expression. Other promoters with similar constitutive activity include, but are not limited to the RSV promoter, and the S V40 promoter. Mammalian promoters such as the beta-actin promoter, ubiquitin C promoter, elongation factor

1 apromoter, tubulin promoter, etc. , may also be used.

[0194] The foregoing SIN configurations are illustrative and non-limiting.

Numerous SIN configurations are known to those of skill in the art. As indicated above, in certain embodiments, the LTR transcription is reduced by about 95% to about 99%. In certain embodiments LTR may be rendered at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95% at least about 96%, at least about 97%, at least about 98%, or at least about 99% transcriptionally inactive.

Insulator element

[0195] In certain embodiments, to further enhance biosafety, insulators are inserted into the LV described herein. Insulators are DNA sequence elements present throughout the genome. They bind proteins that modify chromatin and alter regional gene expression. The placement of insulators in the vectors described herein offer various potential benefits including, inter alia: 1) Shielding of the vector from positional effect variegation of expression by flanking chromosomes (i.e., barrier activity); and 2) Shielding flanking chromosomes from insertional /raw-activation of gene expression by the vector (enhancer blocking). Thus, insulators can help to preserve the independent function of genes or transcription units embedded in a genome or genetic context in which their expression may otherwise be influenced by regulatory signals within the genome or genetic context (see, e.g., Burgess-Beusse et al (2002) Proc. Natl Acad. Sci. USA, 99: 16433; and Zhan et al (2001 ) Hum. Genet., 109: 471). In the present context insulators may contribute to protecting lenti virus-expressed sequences from integration site effects, which may be mediated by cis-acting elements present in genomic DNA and lead to deregulated expression of transferred sequences. In various embodiments LVs are provided in which an insulator sequence is inserted into one or both LTRs or elsewhere in the region of the vector that integrates into the cellular genome.

[0196] The first and best characterized vertebrate chromatin insulator is located within the chicken β-globin locus control region. This element, which contains a DNase-I hypersensitive site-4 (cHS4), appears to constitute the 5' boundary of the chicken β-globin locus (Prioleau et al. (1999) EMBOJ. 18: 4035-4048). A 1.2-kb fragment containing the cHS4 element displays classic insulator activities, including the ability to block the interaction of globin gene promoters and enhancers in cell lines (Chung et al. (1993) Cell, 74: 505-514), and the ability to protect expression cassettes in Drosophila (Id.), transformed cell lines (Pikaart et al. (1998) Genes Dev. 12: 2852-2862), and transgenic mammals (Wang et al. (1997) Nat. Biotechnol, 15: 239-243; Taboit-Dameron et al. (1999) Transgenic Res., 8: 223-235) from position effects. Much of this activity is contained in a 250-bp fragment. Within this stretch is a 49-bp cHS4 core (Chung et al. (1997) Proc. Natl. Acad. Sci., USA, 94: 575-580) that interacts with the zinc finger DNA binding protein CTCF implicated in enhancer-blocking assays (Bell et al. (1999) Cell, 98: 387-396).

[0197] One illustrative and suitable insulator is FB (FH/BEAD-A), a 77 bp insulator element, that contains the minimal CTCF binding site enhancer-blocking components of the chicken β-globin 5' HS4 insulators and a homologous region from the human T-cell receptor alpha/delta blocking element alpha/delta I (BEAD-I) insulator described by Ramezani et al. (2008) Stem Cell 26: 3257-3266. The FB "synthetic" insulator has full enhancer blocking activity. This insulator is illustrative and non-limiting. Other suitable insulators may be used including, for example, the full length chicken beta-globin HS4 or insulator sub-fragments thereof, the ankyrin gene insulator, and other synthetic insulator elements.

Packaging signal.

[0198] In various embodiments the vectors described herein further comprise a packaging signal. A "packaging signal," "packaging sequence," or "psi sequence" is any nucleic acid sequence sufficient to direct packaging of a nucleic acid whose sequence comprises the packaging signal into a retroviral particle. The term includes naturally occurring packaging sequences and also engineered variants thereof. Packaging signals of a number of different retroviruses, including lentiviruses, are known in the art.

Rev Responsive Element (RRE).

[0199] In certain embodiments the LVs described herein comprise a Rev response element (RRE) to enhance nuclear export of unspliced RNA. RREs are well known to those of skill in the art. Illustrative RREs include, but are not limited to RREs such as that located at positions 7622-8459 in the HIV NL4-3 genome (Genbank accession number AF003887) as well as RREs from other strains of HIV or other retroviruses. Such sequences are readily available from Genbank or from the database with URL hiv-web.lad.gov/content/index. Central PolvPurlne Tract (cPPT).

[0200] In various embodiments the vectors described herein further include a central polypurine tract Insertion of a fragment containing the central polypurine tract (cPPT) in lenti viral (e.g., HIV-1) vector constructs is known to enhance transduction efficiency drastically, reportedly by facilitating the nuclear import of viral cDNA through a central DNA flap.

Expression-Stimulating Posttranscriptional Regulatory Element (PRE)

[0201] In certain embodiments the LVs described herein may comprise any of a variety of posttranscriptional regulatory elements (PREs) whose presence within a transcript increases expression of the heterologous nucleic acid (e.g., βAS3) at the protein level.

PREs may be particularly useful in certain embodiments, especially those that involve lenti viral constructs with modest promoters.

[0202] One type of PRE is an intron positioned within the expression cassette, which can stimulate gene expression. However, introns can be spliced out during the life cycle events of a lentivirus. Hence, if introns are used as PRE's they are typically placed in an opposite orientation to the vector genomic transcript

[0203] Posttranscriptional regulatory elements that do not rely on splicing events offer the advantage of not being removed during the viral life cycle. Some examples are the posttranscriptional processing element of herpes simplex virus, the posttranscriptional regulatory element of the hepatitis B virus (HPRE) and the woodchuck hepatitis virus (WPRE). Of these the WPRE is typically preferred as it contains an additional cis-acting element not found in the HPRE. This regulatory element is typically positioned within the vector so as to be included in the RNA transcript of the transgene, but outside of stop codon of the transgene translational unit.

[0204] The WPRE is characterized and described in U.S. Pat. No: 6,136,597. As described therein, the WPRE is an RNA export element that mediates efficient transport of RNA from the nucleus to the cytoplasm. It enhances the expression of transgenes by insertion of a cw-acting nucleic acid sequence, such that the element and the transgene are contained within a single transcript. Presence of the WPRE in the sense orientation was shown to increase transgene expression by up to 7- to 10-fold. Retroviral vectors transfer sequences in the form of cDNAs instead of complete mtron-containing genes as introns are generally spliced out during the sequence of events leading to the formation of the retroviral particle. Introns mediate the interaction of primary transcripts with the splicing machinery. Because the processing of RNAs by the splicing machinery facilitates their cytoplasmic export, due to a coupling between the splicing and transport machineries, cDNAs are often inefficiently expressed. Thus, the inclusion of the WPRE in a vector results in enhanced expression of transgenes.

Transduced Host Cells and Methods of cell transduction.

[0205] The recombinant LV and resulting virus described herein are capable of transferring a nucleic acid {e.g., a nucleic acid encoding an anti-sickling β-globin) sequence into a mammalian cell. For delivery to cells, vectors of the present invention are preferably used in conjunction with a suitable packaging cell line or co-transfected into cells in vitro along with other vector plasmids containing the necessary retroviral genes (e.g., gag and pol) to form replication incompetent virions capable of packaging the vectors of the present invention and infecting cells.

[0206] The recombinant LVs and resulting virus described herein are capable of transferring a nucleic acid (e.g., a nucleic acid encoding an anti-sickling β-globin) sequence into a mammalian cell. For delivery to cells, vectors of the present invention are preferably used in conjunction with a suitable packaging cell line or co-transfected into cells in vitro along with other vector plasmids containing the necessary retroviral genes (e.g, gag and pol) to form replication incompetent virions capable of packaging the vectors of the present invention and infecting cells.

[0207] Typically, the vectors are introduced via transfection into the packaging cell line. The packaging cell line produces viral particles that contain the vector genome.

Methods for transfection are well known by those of skill in the art. After cotransfection of the packaging vectors and the transfer vector to the packaging cell line, the recombinant virus is recovered from the culture media and tittered by standard methods used by those of skill in the art Thus, the packaging constructs can be introduced into human cell lines by calcium phosphate transfection, lipofection or electroporation, generally together with a dominant selectable marker, such as neomycin, DHFR, Glutamine synthetase, followed by selection in the presence of the appropriate drug and isolation of clones. In certain embodiments the selectable marker gene can be linked physically to the packaging genes in the construct

[0208] Stable cell lines wherein the packaging functions are configured to be expressed by a suitable packaging cell are known (see, e.g., U.S. Patent No. 5,686,279, which describes packaging cells). In general, for the production of virus particles, one may employ any cell that is compatible with the expression of lentiviral Gag and Pol genes, or any cell that can be engineered to support such expression. For example, producer cells such as 293T cells and HT1080 cells may be used.

[0209] The packaging cells with a lentiviral vector incorporated in them form producer cells. Producer cells are thus cells or cell-lines that can produce or release packaged infectious viral particles carrying the therapeutic gene of interest (e.g., modified β-globin). These cells can further be anchorage dependent which means that these cells will grow, survive, or maintain function optimally when attached to a surface such as glass or plastic. Some examples of anchorage dependent cell lines used as lentiviral vector packaging cell lines when the vector is replication competent are HeLa or 293 cells and PERC.6 cells.

[0210] Accordingly, in certain embodiments, methods are provided of delivering a gene to a cell which is then integrated into the genome of the cell, comprising contacting the cell with a virion containing a lentiviral vector described herein. The cell (e.g. , in the form of tissue or an organ) can be contacted (e.g., infected) with the virion ex vivo and then delivered to a subject (e.g., a mammal, animal or human) in which the gene (e.g., anti- sickling β-globin) will be expressed. In various embodiments the cell can be autologous to the subject (i.e., from the subject) or it can be non-autologous (i.e., allogeneic or xenogenic) to the subject. Moreover, because the vectors described herein are capable of being delivered to both dividing and non-dividing cells, the cells can be from a wide variety including, for example, bone marrow cells, mesenchymal stem cells (e.g., obtained from adipose tissue), and other primary cells derived from human and animal sources. Alternatively, the virion can be directly administered in vivo to a subject or a localized area of a subject (e.g., bone marrow).

[0211] Of course, as noted above, the lentivectors described herein will be particularly useful in the transduction of human hematopoietic progenitor cells or a hematopoietic stem cells, obtained either from the bone marrow, the peripheral blood or the umbilical cord blood, as well as in the transduction of a CD4 + T cell, a peripheral blood B or T lymphocyte cell, and the like. In certain embodiments particularly preferred targets are CD34 + cells.

Gene therapy.

[0212] In still other embodiments, the present invention is directed to a method for transducing a human hematopoietic stem cell comprising contacting a population of human cells that include hematopoietic stem cells with one of the foregoing lentivectors under conditions to effect the transduction of a human hematopoietic progenitor cell in said population by the vector. The stem cells may be transduced in vivo or in vitro, depending on the ultimate application. Even in the context of human gene therapy, such as gene therapy of human stem cells, one may transduce the stem cell in vivo or, alternatively, transduce in vitro followed by infusion of the transduced stem cell into a human subject In one aspect of this embodiment, the human stem cell can be removed from a human, e.g., a human patient, using methods well known to those of skill in the art and transduced as noted above. The transduced stem cells are then reintroduced into the same or a different human.

Stem cell/progenitor cell gene therapy.

[0213] In various embodiments the lentivectors described herein are particularly useful for the transduction of human hematopoietic progenitor cells or haematopoietic stem cells (HSCs), obtained either from the bone marrow, the peripheral blood or the umbilical cord blood, as well as in the transduction of a CD4 + T cell, a peripheral blood B or T lymphocyte cell, and the like. In certain embodiments particularly preferred targets are CD34 + cells.

[0214] When cells, for instance CD34 + cells, dendritic cells, peripheral blood cells or tumor cells are transduced ex vivo, the vector particles are incubated with the cells using a dose generally in the order of between 1 to SO multiplicities of infection (ΜΟΓ) which also corresponds to 1 x 10 5 to 50 * 10 5 transducing units of the viral vector per 10 s cells. This can include amounts of vector corresponding to 1 , 2, 3, 4, S, 6, 7, 8, 9, 10, IS, 20, 25, 30, 35, 40, 45, and 50 MOI. Typically, the amount of vector may be expressed in terms of HeLa transducing units (TU).

[0215] In certain embodiments cell-based therapies involve providing stem cells and/or hematopoietic precursors, transduce the cells with the lenti virus encoding, e.g. , an anti-sickling human β-globin, and then introduce the transformed cells into a subject in need thereof (e.g., a subject with the sickle cell mutation).

[0216] In certain embodiments the methods involve isolating population of cells, e.g., stem cells from a subject, optionally expand the cells in tissue culture, and administer the lentiviral vector whose presence within a cell results in production of an anti-sickling β- globin in the cells in vitro. The cells are then returned to the subject, where, for example, they may provide a population of red blood cells that produce the anti-sickling β globin.

[0217] In some embodiments of the invention, a population of cells, which may be cells from a cell line or from an individual other than the subject, can be used. Methods of isolating stem cells, immune system cells, etc., from a subject and returning them to the subject are well known in the art. Such methods are used, e.g., for bone marrow transplant, peripheral blood stem cell transplant, etc., in patients undergoing chemotherapy.

[0218] Where stem cells are to be used, it will be recognized that such cells can be derived from a number of sources including bone marrow (BM), cord blood (CB) CB, mobilized peripheral blood stem cells (mPBSC), and the like. In certain embodiments the use of induced pluripotent stem cells (IPSCs) is contemplated. Methods of isolating hematopoietic stem cells (HSCs), transducing such cells and introducing them into a mammalian subject are well known to those of skill in the art.

[0219] In certain embodiments a lentiviral vector described herein (see, e.g„ Figures 15 A, and 16A) is used in stem cell gene therapy for SCD by introducing the βAS3 anti- sickling beta-globin gene into the bone marrow stem cells of patients with sickle cell disease followed by autologous transplantation.

Direct introduction of vector.

[0220] In certain embodiments direct treatment of a subject by direct introduction of the vectors) described herein is contemplated. The lentiviral compositions may be formulated for delivery by any available route including, but not limited to parenteral (e.g., intravenous), intradermal, subcutaneous, oral (e.g., inhalation), transdermal (topical), transmucosal, rectal, and vaginal. Commonly used routes of delivery include inhalation, parenteral, and transmucosal.

[0221] In various embodiments pharmaceutical compositions can include an LV in combination with a pharmaceutically acceptable carrier. As used herein the language "pharmaceutically acceptable carrier" includes solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like, compatible with pharmaceutical administration. Supplementary active compounds can also be incorporated into the compositions.

[0222] In some embodiments, active agents, i.e., a lentiviral described herein and/or other agents to be administered together the vector, are prepared with carriers that will protect the compound against rapid elimination from the body, such as a controlled release formulation, including implants and microencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Methods for preparation of such compositions will be apparent to those skilled in the art. Suitable materials can also be obtained commercially from Alza Corporation and Nova Pharmaceuticals, Inc.

Liposomes can also be used as pharmaceutically acceptable carriers. These can be prepared according to methods known to those skilled in the art, for example, as described in U.S. Pat. No.4,522,811. In some embodiments the composition is targeted to particular cell types or to cells that are infected by a virus. For example, compositions can be targeted using monoclonal antibodies to cell surface markers, e.g., endogenous markers or viral antigens expressed on the surface of infected cells.

[0223] It is advantageous to formulate compositions in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the subject to be treated; each unit comprising a predetermined quantity of a LV calculated to produce the desired therapeutic effect in association with a pharmaceutical carrier.

[0224] A unit dose need not be administered as a single injection but may comprise continuous infusion over a set period of time. Unit dose of the LV described herein may conveniently be described in terms of transducing units (T.U.) of lentivector, as defined by titering the vector on a cell line such as HeLa or 293. In certain embodiments unit doses can range from 10 3 , 10 4 , 10 5 , 10 6 , 10 7 , 10 8 , 10 9 , 10 10 , 10 11 , 10 12 , 10 13 T.U. and higher. [0225] Pharmaceutical compositions can be administered at various intervals and over different periods of time as required, e.g., one time per week for between about 1 to about 10 weeks; between about 2 to about 8 weeks; between about 3 to about 7 weeks; about 4 weeks; about 5 weeks; about 6 weeks, etc. It may be necessary to administer the therapeutic composition on an indefinite basis. The skilled artisan will appreciate that certain factors can influence the dosage and timing required to effectively treat a subject, including but not limited to the severity of the disease or disorder, previous treatments, the general health and/or age of the subject, and other diseases present. Treatment of a subject with a LV can include a single treatment or, in many cases, can include a series of treatments.

[0226] Exemplary doses for administration of gene therapy vectors and methods for determining suitable doses are known in the art It is furthermore understood that appropriate doses of a LV may depend upon the particular recipient and the mode of administration. The appropriate dose level for any particular subject may depend upon a variety of factors including the age, body weight, general health, gender, and diet of the subject, the time of administration, the route of administration, the rate: of excretion, other administered therapeutic agents, and the like.

[0227] In certain embodiments lenti viral gene therapy vectors can be delivered to a subject by, for example, intravenous injection, local administration, or by stereotactic injection (see, e.g., Chen et al. (1994) Proc. Natl. Acad. Sci. USA, 91 : 3054). In certain embodiments vectors may be delivered orally or inhalationally and may be encapsulated or otherwise manipulated to protect them from degradation, enhance uptake into tissues or cells, etc. Pharmaceutical preparations can include a LV in an acceptable diluent, or can comprise a slow release matrix in which a LV is imbedded. Alternatively or additionally, where a vector can be produced intact from recombinant cells, as is the case for retroviral or lenti viral vectors as described herein, a pharmaceutical preparation can include one or more cells which produce vectors. Pharmaceutical compositions comprising a LV described herein can be included in a container, pack, or dispenser, optionally together with instructions for administration.

[0228] The foregoing compositions, methods and uses are intended to be illustrative and not limiting. Using the teachings provided herein other variations on the compositions, methods and uses will be readily available to one of skill in the art. EXAMPLES

[0229] The following examples are offered to illustrate, but not to limit the claimed invention.

Example 1

Development of Next Generation Vectors for Gene Therapy of Sickle Cell Disease

[0230] Autologous hematopoietic stem cell gene therapy is an approach to treating sickle cell disease (SCD) patients that may result in lower morbidity than allogeneic transplantation. We previously examined the potential of a LV (CCL-βAS3-FB) encoding a human beta-globin (HBB) gene engineered to impede sickle hemoglobin polymerization (HBBAS3) to transduce human BM CD34 + cells from SCD donors and prevent sickling of rbc produced by in vitro differentiation {see, e.g., U.S. Patent Pub. No: 2015/0224209; PCT Pub. No: WO 2014/043131; Romero etal. (2013)J. Clin. Invest, 128(8): 3317-3330)). The CCL-βAS3-FB LV transduced BM CD34 + cells from either healthy or SCD donors at similar levels, based on quantitative PCR and colony-forming unit progenitor analysis. Consistent expression of HBB AS3 mRNA and HbAS3 protein compromised a fourth of the total β-globin-like transcripts and Hb tetramers. Upon deoxygenation, a lower percentage of HBBAS3-transduced rbc exhibited sickling compared with mock-transduced cells from sickle donors. Transduced BM CD34 + cells were transplanted into immunodeficient mice, and the human cells recovered after 2-3 months were cultured for erythroid differentiation, which showed levels of HBBAS3 mRNA similar to those seen in the CD34 + cells that were directly differentiated in vitro. These results demonstrated that the CCL-βAS3-FB LV is capable of efficient transfer and consistent expression of an effective anti-sickling β-globin gene in human SCD BM CD34 + progenitor cells, improving physiologic parameters of the resulting rbc.

[0231] In an effort to further improve vector performance, we initially evaluated the

CCL-βAS3-FB construct {see, Fig. 1 A) and compared it to the smaller CCL-GLOBEl- βAS3 construct (see, Fig. IB; Miccio et a/.(2008) Proc. Natl. Acad. Sci. USA, 10547- 10552). In particular, raw titers were evaluated as well as the erythroid dose response and myeloid dose response (see, Fig. IB). CCL-βAS3-FB showed a significantly lower average raw titer (Fig. IB, top panel), however, erythroid dose response (Fig. IB, middle panel) was marginally higher for GLOBE1-βAS3 and the myeloid dose response was essentially the same for both constructs (Fig. IB, bottom panel). [0232] To investigate whether overall vector size impacts titer or gene transfer more than the presence of discrete sequences, a series of CCLc-βAS3-FB derivative were designed as illustrated in Figure 2. These included, but were not limited to deletions of HS4 (e.g, CCLc-βAS3-FB(ΔHS4), elimination of WPRE (e.g., CCLc-βAS3-FB-(ΔWPRE), deletions of both HS4 and WPRE (e.g., CCLc-βAS3-FB-(ΔHS4/AWPRE), deletion of HS4 and insertion of a spacer element therefor (e.g., CCLc-βAS3-FB-(ΔHS4)-(+HPRT), elimination of IVS2 (e.g., CCLc-βAS3-FB-(ΔIVS2), and the like.

[0233] Figure 3 A shows the average fold difference in titers for various ΔHS4 constructs. As shown, reduction of length via removal of HS4 improves titer. Figure 3B shows the effect of vector length on gene transfer to HSC. As shown, a reduction in vector length improves gene transfer as well. Figure 3C shows the effect of vector length (CCLc- βAS3-FB versus CCLc-βAS3-(ΔHS4) on expression level. As shown while a reduction in vector length improves titer and gene transfer, removal of HS4 impaired expression.

[0234] In conclusion it appears that reduction of vector length through deletion of HS4 improved tier and gene transfer, but impaired expression. Moreover, replacing HS4 with HPRT sniffer (l.lkb) did not improve titer or gene transfer suggesting that size is the predominant factor influencing titer and perhaps gene transfer.

[0235] To further explore this, we compared the performance of three constructs of different length: 1) CCLc-βAS3-FB (8.9kb), 2) CCLc-βAS3-FB-(ΔHS4) (6.3 kb), and 3) CCL-GLOBE1-AS3 (6.3 kb). The structures of these three constructs are illustrated in Figure 4.

[0236] The titer of each of these vectors is shown in Figure SA, while transduction levels are shown in Figure SB, and expression levels in Figure SC. Expression levels are also summarized below in Table 1.

Table 1. Expression levels of CCLc-βAS3-FB (8.9kb), CCLc-βAS3-FB-(ΔHS4) (6.3 kb), and CCL-GLOBE 1 - AS3 (6.3 kb).

[0237] Figure 6 A shows the raw titers plotted as a function of construct size for IS different lenti viral constructs. Fig. 6B shows the expression levels (vector copy number (VCN)) plotted as a function of construct size for these different lentiviral constructs.

[0238] As can be seen, reduction of vector length through removal of discrete sequences improves titer and gene transfer. However certain elements must be preserved to maintain erythroid specific expression. The challenge was to identify the best combination of elements that offer high-level and erythroid specific expression within the smallest footprint.

[0239] We engineered optimized derivatives of CCLc-βAS3-FB, that are capable of driving lineage-restricted expression of an anti- sickling β-globin like gene (βAS3) (see, e.g., Figures 8B, 15A, 16A). Creation of the "optimized mini-LCR", present within the derivative constructs, occurred through redefining the putative boundaries of the LCR's HS core sequences using published genomic data available through ENCODE (Accessible via the UCSC Genome Browser). Specifically, the "Open Chromatin" track sets, which combine DNasel hypersensitivity, Formaldehyde- Assisted Isolation of Regulatory

Elements, and chromatin immunoprecipitation data to identify accessible chromatin regions, was combined with the "transcription Factor ChlP-seq" and "histone modification" track sets to generate correct boundaries for the LCR's HS core sequences (see, e.g., Figure 7).

[0240] These novel defined LCR HS core sequences (HS2(~420 bp), HS3(~340bp), HS4(~410 bp)) were then used to replace the putative LCR HS sequences present within the "mini-LCR" (~3.6 kb reduced to ~1.2 kb) to produce an "optimized mini-LCR" (see, e.g., Figure 8A).

[0241] In addition, we added elements to the "optimized mini-LCR" known to facilitate position independent expression of β-globin such as the murine GATA1-HS2 (~220 bp) and/or the human Ankyrin insulator (~150 bp) elements (see, e.g., Figures 8B, and 10).

[0242] Based on the vector length-titer relationship shown in Figure 9, it was predicted that a reduction in vector length of about 2-3kb should increase titer about 3-fold.

[0243] These vectors, rationally-designed for reduced sizes of the LCR fragments and added transcriptional enhancing elements may be produced at higher titers than the original β-globin lentiviral vector and have improved gene transfer to human HSC while retaining strong erythroid-specific gene expression. [0244] Figure 15 A illustrates the pCCLC-βAS3-FB(+CoreLCR w-Ank-R) optimized vector ((10528 bp), while Figure 15B illustrates the nucleic acid sequence of the vector. Regions of interest in this vector are shown below in Table 2.

Table 2. Locations of various regions of interest in the pCCLC-βAS3-FB(+CoreLCR w- Ank-R) optimized vector ((10528 bp).

[0245] Figure 16A illustrates the pCCL-βAS3-(-+mGATA-HS2-Ank-dIVS2-

CoreLCRopti)-FB optimized vector (10483 bp), while Figure 16B illustrates the nucleic acid sequence of the vector. Regions of interest in this vector are shown below in Table 2.

Table 3. Locations of various regions of interest in the pCCL-βAS3-(+mGATA-HS2-Ank- dIVS2-CoreLCRopti)-FB optimized vector.

[0246] Raw titer data for CCLc-βAS3-(+Ank)-(CoreLCR)-(Opti.)-FB are shown in

Table 4 and Figure 11.

Table 4. Raw titer data.

[0247] Average raw titer data CCLc-βAS3-(+Ank)-(CoreLCR)-(Opti.)-FB normalized to BAS3-FB are shown in Figure 12, top panel, while expression data are shown in Figure 12, bottom panel. CCL-GLOBE-FB expression (Avg % AS3) significantly lower than CCLc-B AS3-FB. CCLc-ANK-CORELCR-Opti expression not significantly different than CCLc-BAS3-FB.

[0248] Figure 13 shows that replacing the LCR with ANK and optimized HS2, HS3, and HS4 core element increased titer by three fold. Figure 14 and Table S shows that replacing the LCR with ANK and only core elements achieves levels of expression similar to CCLc-βAS3-FB(ΔHS4).

Table 5. Construct expression levels.

F.Yflmple 2

Oprimized Derivatives of CCLc-B AS3-FB

[0249] We have engineered optimized derivatives of "CCLc-βAS3-FB", which are capable of driving lineage-restricted expression of an anti-sickling β-globin like gene

(βAS3). Creation of the "optimized mini-LCR", present within the derivative constructs, occurred through redefining the putative boundaries of the LCR's HS core sequences using published genomic data available through ENCODE (Accessible via the UCSC Genome Browser). Specifically, the "Open Chromatin" track sets, which combine DNasel hypersensitivity, Formaldehyde-Assisted Isolation of Regulatory Elements, and chromatin immunoprecipitation data to identify accessible chromatin regions, was combined with the "transcription Factor ChlP-seq" and "histone modification" track sets to generate correct boundaries for the LCR's HS core sequences.

[0250] These novel defined LCR HS core sequences (HS2(~420 bp), HS3(~340bp),

HS4(~410 bp)) were then used to replace the putative LCR HS sequences present within the " mini-LCR" (-3.6 Kb reduced to ~12 Kb) to produce an "optimized mini-LCR" (see, e.g., Figure 17).

[0251 ] In addition, we added elements to the "optimized mini-LCR" known to facilitate position independent expression of β-globin such as the murine mGATA 1 -HS2 (-220 bp) and/or the Human Ankyrin Insulator (-150 bp) elements (see, e.g., Figures 18A and 18B). [0252] We also tested the mGATAl-HS2 and the human ankyrin insulator elements in alternative configurations in combination with the "optimized mini-LCR" (see, e.g., Figure 18C).

[0253] We also tested the murine mGATAl-HS2 and the human ankyrin insulator elements with full-length erythroid enhancers (HS2 and HS3) (see, e.g., Figure 19).

[0254] These vectors, rationally-designed for reduced sizes of the LCR fragments and added transcriptional enhancing elements may be produced at higher titers than the original β-globin lentiviral vector ("CCLc-βAS3-FB") and possess improved gene transfer to human HSPCs while retaining erythroid-specific gene expression.

[0255] As illulstrated in Figure 20, we observed that deletion of full-length HS4

(leaving HS 2 and 3 in place, as in GLOBE vectors) and partial deletion of intron-2 sequences from "CCLc-βAS3-FB" increased titer 2.8-fold (see "CCLc-βAS3-FB - ΔHS4/ΔIVS2"). Addition of mGATA/ANK element to "CCLc-βAS3-FB -ΔHS4/AIVS2" did not overtly negate gains in titer achieved from removal of sequence from "CCLc-βAS3- FB" (see "CCLc-βAS3-FB -mGATA/ANK -ΔHS4/MVS2"). Replacement of full-length HS2 and HS3 elements in "CCLc-βAS3-FB -mGATA/ANK -ΔHS4/ΔIVS2" with

"CoreLCR Opti" increased raw titer 4.3-fold when compared to parental (see "CCLc-βAS3- FB -mGATA/ANK -CoreLCR Opti -ΔHS234/ΔIVS2"). Addition of intron-2 sequence and replacement of mGATA/ANK with only ANK (and placed in an alternative location) did not overtly negate gains in titer achieved from size reduction (see "CCLc-βAS3-FB -

CoreLCR Opti -ANK"). These data show that improved raw titer can be achieved through deletion of full-length HS elements and inclusion of core elements with mGATA-HS2 and/or ANK.

[0256] Myeloid gene transfer studies typically reflect the level of gene marking seen in Human bone marrow following gene therapy. As shown in Figure 21 , we observed that deletion of full-length HS4 and partial deletion of intron-2 sequences from "CCLc-βAS3- FB" increased gene transfer -1.8, ~1.7, or ~1.8-fold when CD34+ HSPCs were transduced at le6, le7, or le8 (TU/ml), respectively (see "CCLc-βAS3-FB -ΔHS4/ΔIVS2"). Addition of mGATA/ANK element to "CCLc-βAS3-FB -ΔHS4/ΔIVS2" increased gene transfer -2.0, -1.6, or ~1.6-fold when CD34+ HSPCs were transduced at le6, le7, or le8 (TU/ml), respectively (see "CCLc-βAS3-FB -mGATA/ANK -ΔHS4/ΔIVS2"). Replacement of full- length HS2 and HS3 elements present within "CCLc-βAS3-FB -mGATA/ANK - ΔHS4/ΔIVS2" with "CoreLCR Opti" increased gene transfer -2.4, -2.1, or ~2.5-fold when CD34+ HSPCs were transduced at 1 x 106, 1 x 107, or 1 x 108 (TU/ml), respectively (see "CCLc-βAS3-FB -mGATA/ANK -CoreLCR Opti -ΔHS234/ATVS2"). Addition of intron-2 sequence and replacement of mGATA/ANK with only ANK (and placed in an alternative location) increased gene transfer ~2.1, ~1.9, or ~2.0-fold when CD34+ HSPCs were transduced at 1 x 10 6 , 1 x 10 7 , or 1 x 10 8 (TU/ml), respectively (see "CCLc-βAS3-FB - CoreLCR Opti -ANK"). These data show that improved gene transfer to CD34+ HSPCs can be achieved through deletion of full-length HS elements and inclusion of "CoreLCR Opti" and mGATA-HS2 and/or ANK.

[0257] Erythroid expression studies offer insight into the total level of expression that can be achieved globally (%transgene/ (%transgene + total β-globin "like" transcripts)) and can be further normalized to vector copy number to estimate the level of expression per vector insertion. As shown in Figure 22, we observed that deletion of full-length HS4 and partial deletion of intron-2 sequences from "CCLc-βAS3-FB" increased total transgene expression by ~1.5-fold by way of increased gene transfer to target cells. When expression was normalized to VCN, total transgene expression was ~0.9-fold that of the parental vector (see "CCLc-βAS3-FB -ΔHS4/ΔIVS2"). Addition of mGATA/ANK element to "CCLc- βAS3-FB -ΔHS4/ΔIVS2" increased total expression by ~2-fold when compared to parental. The increase in total expression results from synergy between greater gene transfer to target cells and higher expression per VCN when compared to parental (see "CCLc-βAS3-FB - mGATA/ANK -ΔHS4/ΔIVS2"). Replacement of full-length HS2 and HS3 elements present within "CCLc-βAS3-FB -mGATA/ANK -ΔHS4/ΔIVS2" with "CoreLCR Opti" resulted in comparable total transgene expression when compared to "CCLc-βAS3-FB" by way of increased gene transfer to target cells. When expression was normalized to VCN, total transgene expression was ~0.6-fold that of the parental vector (see "CCLc-βAS3-FB - mGATA/ANK -CoreLCR Opti -ΔHS234/ATVS2").

[0258] As illustrated in Figure 23, when HS4, HS3, and HS2 were replaced with

"CoreLCR Opti" alone, gene transfer was improved by ~3-fold when CD34+ HSPCs were transduced at 1 x 10 7 (TU/ml) (see "CCLc-βAS3-FB -CoreLCR Opti"). The addition of ANK to "CoreLCR Opti" did not overtly negate improvements in gene transfer gained by removing HS4, HS3, and HS2 elements (see "CCLc-βAS3-FB -CoreLCR Opti -Ank").

The addition of mGATA/ANK to "CoreLCR Opti" (now, same location as ANK in "CCLc- βAS3-FB -CoreLCR Opti -ANK"), resulted in a ~3-fold improvement in gene transfer (see "CCLc-βAS3-FB -CoreLCR Opti -mGATA/ANK"). Replacement of HS4, HS3, and HS2 with any "CoreLCR Opti" variant did not result in any detectable expression in CD34+ HSPCs cultured under myeloid culture conditions. When this finding is taken together with other expression data supplied herein, it implies that all "CoreLCR Opti" variants retain lineage restricted expression. Addition of intron-2 sequence and replacement of mGATA/ANK with only ANK (and placed in an alternative location) resulted in ~1.5-fold increased total transgene expression by way of increased gene transfer to target cells (see "CCLc-βAS3-FB -CoreLCR Opti -ANK"). These data show that, 1). Addition of mGATA/ANK element to "CCLc-βAS3-FB -ΔHS4/ΔIVS2" improved enhancer output, 2). "CCLc-βAS3-FB -mGATA/ANK -CoreLCR Opti -ΔHS234/ΔIVS2" or "CCLc-βAS3-FB - CoreLCR Opti -ANK" provide similar or superior total transgene expression when compared to parental.

[0259] As shown in Figure 24, when HS4, HS3, and HS2 were replaced with

"CoreLCR Opti" alone or in combination with mGATA/ANK or only ANK, total expression was comparable or superior to "CCLc-βAS3-FB" by way of increased gene transfer to target cells. This observation can be made at the 6.6 x 10 6 or 1 x 10 7 (TU/ml) transduction conditions. When mGATA/ANK was placed adjacent to HS2 (now, same location as ANK in "CCLc-βAS3-FB -CoreLCR Opti -ANK"), increased total comparable or superior total transgene expression can be observed. This observation, when taken together with those stated herein, suggest that mGATA/ANK can work in alternate configurations (compare "CCLc-βAS3-FB -CoreLCR Opti -mGATA/ANK" to "CCLc- βAS3-FB -mGATA/ANK -CoreLCR Opti -ΔHS234/ΔIVS2").

[0260] It is understood that the examples and embodiments described herein are for illustrative purposes only and that various modifications or changes in tight thereof will be suggested to persons skilled in the art and are to be included within the spirit and purview of this application and scope of the appended claims. All publications, patents, and patent applications cited herein are hereby incorporated by reference in their entirety for all purposes.

SEQUENCE LISTING

Sequence ID No:l pC(XC-βAS3-FB(+CoreLCR w-Ank-R) optimized vector (10528 bp)

atttttacgg ttcctggcct tttgctggcc ttttgctcac atgttctttc ctgcgttatc 60 ccctgattct gtggataacc gtattaccgc ctttgagtga gctgataccg ctcgccgcag 120 ccgaaegacc gagcgcagcg agtcagtgag cgaggaagcg gaagagcgcc caatacgcaa 180 accgcctctc cccgcgcgtt ggccgattca ttaatgcagc tggcacgaca ggtttcccga 240 ctggaaagcg ggcagtgagc gcaacgcaat taatgtgagt tagctcactc attaggcacc 300 ccaggcttta cactttatgc ttccggctcg tatgttgtgt ggaattgtga gcggataaca 360 atttcacaca ggaaacagct atgaccatga ttacgccaag cgcgcaatta accctcacta 420 aagggaacaa aagctggagc tgcaagcttg gccattgcat acgttgtatc catatcataa 480 tatgtacatt tatattggct catgtccaac attaccgcca tgttgacatt gattattgac 540 tagttattaa tagtaatcaa ttacggggtc attagttcat agcccatata tggagttccg 600 cgttacataa cttacggtaa atggcccgcc tggctgaccg cccaacgacc cccgcccatt 660 gacgtcaata atgacgtatg ttcccatagt aacgccaata gggactttcc attgacgtca 720 atgggtggag tatttacggt aaactgccca cttggcagta catcaagtgt atcatatgcc 780 aagtacgccc cctattgacg tcaatgacgg taaatggccc gcctggcatt atgcccagta 840 catgacctta tgggactttc ctacttggca gtacatctac gtattagtca tcgctattac 900 catggtgatg cggttttggc agtacatcaa tgggcgtgga tagcggtttg actcacgggg 960 atttccaagt ctccacccca ttgacgtcaa tgggagtttg ttttggcacc aaaatcaacg 1020 ggactttcca aaatgtcgta acaactccgc cccattgacg caaatgggcg gtaggcgtgt 1080 acggtgggag gtctatataa gcagagctcg tttagtgaac cggggtctct ctggttagac 1140 cagatctgag cctgggagct ctctggctaa ctagggaacc cactgcttaa gcctcaataa 1200 agcttgcctt gagtgcttca agtagtgtgt gcccgtctgt tgtgtgacte tggtaactag 1260 agatccctca gaccctttta gtcagtgtgg aaaatctcta gcagtggcgc ccgaacaggg 1320 acttgaaagc gaaagggaaa ccagaggagc tctctcgacg caggactcgg cttgctgaag 1380 cgcgcacggc aagaggcgag gggcggcgac tggtgagtac gccaaaaatt ttgactagcg 1440 gaggctagaa ggagagagat gggtgcgaga gcgtcagtat taagcggggg agaattagat 1500 cgcgatggga aaaaattcgg ttaaggccag ggggaaagaa aaaatataaa ttaaaacata 1560 tagtatgggc aagcagggag ctagaacgat tcgcagttaa tcctggcctg ttagaaacat 1620 cagaaggctg tagacaaata ctgggacagc tacaaccatc ccttcagaca ggatcagaag 1680 aacttagatc attatataat acagtagcaa ccctctattg tgtgcatcaa aggatagaga 1740 taaaagacac caaggaagct ttagacaaga tagaggaaga gcaaaacaaa agtaagacca 1800 ccgcacagca agcggccgct gatcttcaga cctggaggag gagatatgag ggacaattgg 1860 agaagtgaat tatataaata taaagtagta aaaattgaac cattaggagt agcacccacc 1920 aaggcaaaga gaagagtggt gcagagagaa aaaagagcag tgggaatagg agctttgttc 1980 cttgggttct tgggagcagc aggaagcact atgggcgcag cgtcaatgac gctgacggta 2040 caggccagac aattattgtc tggtatagtg cagcagcaga acaatttgct gagggctatt 2100 gaggcgcaac agcatctgtt gcaactcaca gtctggggca tcaagcagct ccaggcaaga 2160 atcctggctg tggaaagata cctaaaggat caacagctcc tggggatttg ygg Ltgctct 2220 ggaaaactca tttgcaccac tgctgtgcct tggaatgcta gttggagtaa taaatctctg 2280 gaacagattt ggaatcacac gacctggatg gagtgggaca gagaaattaa caattacaca 2340 agcttaatac actccttaat tgaagaatcg caaaaccagc aagaaaagaa tgaacaagaa 2400 ttattggaat tagataaatg ggcaagtttg tggaattggt ttaacataac aaattggctg 2460 tggtatataa aattattcat aatgatagta ggaggcttgg taggtttaag aatagttttt 2520 gctgtacttt ctatagtgaa tagagttagg cagggatatt caccattatc gtttcagacc 2580 cacctcccaa ccccgagggg acccgacagg cccgaaggaa tagaagaaga aggtggagag 2640 agagacagag acagatccat tcgattagtg aacggatctc gacggtatcg atctcgacac 2700 aaatggcagt attcatccac aattttaaaa gaaaaggggg gattgggggg tacagtgcag 2760 gggaaagaat agtagacata atagcaacag acatacaaac taaagaatta caaaaacaaa 2820 ttacaaaaat tcaaaatttt cggg Lttatt acagggacag cagagatcca gtttggg Leg 2880 aggatategg ateggaatte tctagatgat caggatccct cgagccctta tcgatcacga 2940 gactagcctc gactactagt ggagatcccc egggctgeag agecagaage accataaggg 3000 acatgataag ggagecagea gacctctgat ctcttcctga atgetaatet taaacatcct 3060 gaggaagaat gggacttcca tttggggtgg gectatgata gggtaataag acagtagtga 3120 atatcaagct acaaaaagee ccctttcaaa ttcttctcag tcctaacttt tcatactaag 3180 cccagtcctt ccaaagcaga ctgtgaaaga gtgatagttc egggagacta geactgeaga 3240 ttcegggtea ctgtgagtgg gggaggcagg gaagaagggc tcacaggaca gtcaaaccat 3300 gccccctgtt tttccttctt caagtagacc tctataagac aacagagaca actaaggctg 3360 agtggccagg cgaggagaaa ccatctcgcc gtaaaacatg gaaggaacac ttcaggggaa 3420 aggtggtatc tctaagcaag agaactgagt ggagtcaagg ctgagagatg caggataagc 3480 aaatgggtag tgaaaagaca ttcatgagga cagctaaaac aataagtaat gtaaaataca 3540 gcatagcaaa actttaacct ccaaatcaag cctctacttg aatccttttc tgagggatga 3600 ataaggcata ggcatcaggg gctgttgcca atgtgcatta gctgtttgca gcctcacctt 3660 ctttcatgga gtttaagata tagtgtattt tcccaaggtt tgaactagct cttcatttct 3720 ttatgtttta aatgcactga cctcccacat tcccttttta gtaaaatatt cagaaataat 3780 ttaaatacat cattgcaatg aaaataaatg ttttttatta ggcagaatcc agatgctcaa 3840 ggcccttcat aatatccccc agtttagtag ttggacttag ggaacaaagg aacctttaat 3900 agaaattgga cagcaagaaa gcgagcttag tgatacttgt gggccagggc attagccaca 3960 ccagccacca ctttctgata ggcagcctgc actggtgggg tgaattcttt gccaaagtga 4020 tgggccagca cacagaccag cacgttgccc aggagctgtg ggaggaagat aagaggtatg 4080 aacatgatta gcaaaagggc ctagcttgga ctcagaataa tccagcctta tcccaaccat 4140 aaaataaaag cagaatggta gctggattgt agctgctatt agcaatatga aacctcttac 4200 atcagttaca atttatatgc agaaatattt atatgcagaa atattgctat tgccttaacc 4260 cagaaattat cactgttatt ctttagaatg gtgcaaagag gcatgataca ttgtatcatt 4320 attgccctga aagaaagaga ttagggaaag tattagaaat aagataaaca aaaaagtata 4380 ttaaaagaag aaagcatttt ttaaaattac aaatgcaaaa ttaocctgat ttggtcaata 4440 tgtgtaccct gttacttctc cccttcctat gacatgaact taaccataga aaagaagggg 4500 aaagaaaaca tcaagggtcc catagactca ccctgaagtt ctcaggatcc acgtgcagct 4560 tgtcacagtg cagctcactc agctgggcaa aggtgccctt gaggttgtcc aggtgagcca 4620 ggccatcact aaaggcaccg agcactttct tgccatgagc cttcacctta gggttgccca 4680 taacagcatc aggagtggac agatccccaa aggactcaaa gaacctctgg gtccaagggt 4740 agaccaccag cagcctaagg gtgggaaaat agaccaatag gcagagagag tcagtgccta 4800 tcagaaaccc aagagtcttc tctgtcteca catgcccagt ttctattggt ctccttaaac 4860 ctgtcttgta accttgatac caacctgccc agggcctcac caccaacggc atccacgttc 4920 accttgtccc acagggcagt aacggcagac ttctcctcag gagtcaggtg caccatggtg 4980 tctgtttgag gttgctagtg aacacagttg tgtcagaagc aaatgtaagc aatagatggc 5040 tctgccctga cttttatgcc cagccctggc tcctgccctc cctgctcctg ggagtagatt 5100 ggccaaccct agggtgtggc tccacagggt gaggtctaag tgatgacagc cgtacctgtc 5160 cttggctctt ctggcactgg cttaggagtt ggacttcaaa ccctcagccc tccctctaag 5220 atatatctct tggccccata ccatcagtac aaattgctac taaaaacatc ctcctttgca 5280 agtgtattta cccgacgcgt cggcgataag cttgatccat cgatgacgtg cgggccaggc 5340 ccccgagggc cttaacggcc ccagaggcgc ttgctgtcgg gccgggcgct cccggcacgg 5400 gcgggcggag gggtggcgcc cgcctgggga ccgcagatta caagagcacc tcctccccca 5460 accccaggag gccccgctcc ccatacgtat atgtgtatat atatatatat attcaggaaa 5520 taatatattc tagaatatgt cacattctgt ctcaggcatc cattttcttt atgatgccgt 5580 ttgaggtgga gttttagtca ggtggtcage ttctcctttt ttttgccatc tgccctgtaa 5640 gcatcctgct ggggacccag ataggagtca tcactctagg ctgagaacat ctgggcacac 5700 accctaagcc tcagcatgac tcatcatgac tcagcattgc tgtgcttgag ccagaaggtt 5760 tgcttagaag gttacacaga accagaaggc gggggtgggg cactgacccc gacaggggcc 5820 tggccagaac tgctcatgct tggactatgg gaggtcacta atggagacac acagaaatgt 5880 aacaggaact aaggaaaaac tgaagctttg ggggtatagg ggagcagtcc catgtagtag 5940 tagaatgaaa aatgctgcta tgctgtgcct cccccacctt tcccatgtct gccctctact 6000 catggtctat ctctcctggc tcctgggagt catggactcc acccagcacc accaacctga 6060 cctaaccacc tatctgagcc tgccagccta taacccatct gggccctgat agctggtggc 6120 cagccctgac cccaccccac cctccctgga acctctgata gacacatctg gcacaccagc 6180 tcgcaaagtc accgtgaggg tcttgtgttt gctgagtcaa aattccttga aatccaagtc 6240 cttagagact cccaggcttg gattcaaagc tcctgacttt ctgtctagtg tatgtgcagt 6300 gagccccttt tcctctaact gaaagaagga aaaaaaaatg gaacccaaaa tattctacat 6360 agtttccatg tcacagccag ggctgggcag tctcctgtta tttcttttaa aataaatata 6420 tcatttaaat gcataaataa gcaaaccctg ctcgggaatg ggagggagag tctctggagt 6480 ccaccccttc tcggccctgg ctctgcagat agtgctatca aagccctgac agagccctgc 6540 ccattgctgg gccttggagt gagtcagcct agtagagagg cagggcaagc catctcatag 6600 ctgctgagtg ggagagagaa aagggctcat tgtctataaa ctcaggtcat ggctattctt 6660 atggcctact cgaccacgag ggaattccga taatcaacct ctggattaca aaatttgtga 6720 aagattgact ggtattctta actatgttgc tccttttacg ctatgtggat acgctgcttt 6780 aatgcctttg tatcatgcta ttgcttcccg tatggctttc attttctcct ccttgtataa 6840 atcctggttg ctgtctcttt atgaggagtt gtggcccgtt gtcaggcaac gtggcgtggt 6900 gtgcactgtg tttgctgacg caaeccccac tggttggggc attgccacca cctgtcagct 6960 cctttccggg actttcgctt tccccctccc tattgccacg gcggaactca tcgccgcctg 7020 ccttgcccgc tgctggacag gggctcggct gttgggcact gacaattccg tggtgttgtc 7080 ggggaaatca tcgtcctttc cttggctgct cgcctgtgtt gccacctgga ttctgcgcgg 7140 gacgtccttc tgctacgtcc cttcggccct caatccagcg gaccttcctt cccgcggcct 7200 gctgccggct ctgcggcctc ttcegcgtct tcgccttcgc cctcagacga gtcggatctc 7260 cctttgggcc gcctccccgc atcgataccg tcgacctcga gacctagaaa aacatggcca 7320 attcgagctc ggtaccttta agaccaatga cttacaaggc agctgtagat cttagccact 7380 ttttaaaaga aaagggggga ctggaagggc taattcactc ccaacgaaga caagatccca 7440 gggatgtacg tccctaaccc gctagggggc agcacccagg cctgcactgc cgcctgccgg 7500 caggggtcca gtcctgcttt ttgcttgtac tgggtctctc tggttagacc agatctgagc 7560 ctgggagctc tctggctaac tagggaaccc actgcttaag cctcaataaa gcttgccttg 7620 agtgcttcaa gtagtgtgtg cccgtctgtt gtgtgactct ggtaactaga gatccctcag 7680 acccttttag tcagtgtgga aaatctctag cagtagtagt tcatgtcatc ttattattca 7740 gtatttataa cttgcaaaga aatgaatatc agagagtgag aggaacttgt ttattgcagc 7800 ttataatggt tacaaataaa gcaatagcat cacaaatttc acaaataaag catttttttc 7860 actgcattct agttgtggtt tgtccaaact catcaatgta tcttatcatg tctggctcta 7920 gctatcccgc ccctaactcc gcccatcccg cccctaactc cgcccagttc cgcccattct 7980 ccgccccatg gctgactaat tttttttatt tatgcagagg ccgaggccgc ctcggcctct 8040 gagctattcc agaagtagtg aggaggcttt tttggaggcc taggcttttg cgtcgagacg 8100 tacccaattc gccctatagt gagtcgtatt acgcgcgctc actggccgtc gttttacaac 8160 gtcgtgactg ggaaaaccct ggcgttaccc aacttaatcg ccttgcagca catccccctt 8220 tcgccagctg gcgtaatagc gaagaggccc gcaccgatcg cccttcccaa cagttgcgca 8280 gcctgaatgg cgaatggcgc gacgcgccct gtagcggcgc attaagcgcg gcgggtgtgg 8340 tggttacgcg cagcgtgacc gctacacttg ccagcgccct agcgcccgct cctttcgctt 8400 tcttcccttc ctttctcgcc acgttcgccg gctttccccg tcaagctcta aatcgggggc 8460 tccctttagg gttccgattt agtgctttac ggcacctcga ccccaaaaaa cttgattagg 8520 gtgatggttc acgtagtggg ccatcgccct gatagacggt ttttcgccct ttgacgttgg 8580 agtccacgtt ctttaatagt ggactcttgt tccaaactgg aacaacactc aaccctatct 8640 cggtctattc ttttgattta taagggattt tgccgatttc ggcctattgg ttaaaaaatg 8700 agctgattta acaaaaattt aacgcgaatt ttaacaaaat attaacgttt acaatttccc 8760 aggtggcact tttcggggaa atgtgcgcgg aacccctatt tgtttatttt tctaaataca 8820 ttcaaatatg tatccgctca tgagacaata accctgataa atgcttcaat aatattgaaa 8880 aaggaagagt atgagtattc aacatttccg tgtcgccctt attccctttt ttgcggcatt 8940 ttgccttcct gtttttgctc acccagaaac gctggtgaaa gtaaaagatg ctgaagatca 9000 gttgggtgca cgagtgggtt acatcgaact ggatctcaac agcggtaaga tccttgagag 9060 ttttcgcccc gaagaacgtt ttccaatgat gagcactttt aaagttctgc tatgtggcgc 9120 ggtattatcc cgtattgacg ccgggcaaga gcaactcggt cgccgcatac actattctca 9180 gaatgacttg gttgagtact caccagtcac agaaaagcat cttacggatg gcatgacagt 9240 aagagaatta tgcagtgctg ccataaccat gagtgataac actgcggcca acttacttct 9300 gacaacgatc ggaggaccga aggagctaac cgcttttttg cacaacatgg gggatcatgt 9360 aactcgcctt gatcgttggg aaccggagct gaatgaagcc ataccaaacg acgagcgtga 9420 caccacgatg cctgtagcaa tggcaacaac gttgcgcaaa ctattaactg gcgaactact 9480 tactctagct tcccggcaac aattaataga ctggatggag gcggataaag ttgcaggacc 9540 acttctgcgc tcggcccttc cggctggctg gtttattgct gataaatctg gagccggtga 9600 gcgtgggtct cgcggtatca ttgcagcact ggggccagat ggtaagccct cccgtatcgt 9660 agttatctac acgacgggga gtcaggcaac tatggatgaa cgaaatagac agatcgctga 9720 gataggtgcc tcactgatta agcattggta actgtcagac caagtttact catatatact 9780 ttagattgat ttaaaacttc atttttaatt taaaaggatc taggtgaaga tcctttttga 9840 taatctcatg accaaaatcc cttaacgtga gttttcgttc cactgagcgt cagaccccgt 9900 agaaaagatc aaaggatctt cttgagatcc tttttttctg cgcgtaatct gctgcttgca 9960 aacaaaaaaa ccaccgctac cagcggtggt ttgtttgccg gatcaagagc taccaactct 10020 ttttccgaag gtaactggct tcagcagagc gcagatacca aatactgtcc ttctagtgta 10080 gccgtagtta ggccaccact tcaagaactc tgtagcaccg cctacatacc tcgctctgct 10140 aatcctgtta ccagtggctg ctgccagtgg cgataagtcg tgtcttaccg ggttggactc 10200 aagacgatag ttaccggata aggcgcagcg gtcgggctga acggggggtt cgtgcacaca 10260 gcccagcttg gagcgaacga cctacaccga actgagatac ctacagcgtg agctatgaga 10320 aagcgccacg cttcccgaag ggagaaaggc ggacaggtat ccggtaagcg gcagggtcgg 10380 aacaggagag cgcacgaggg agcttccagg gggaaacgcc tggtatcttt atagtcctgt 10440 cgggtttcgc cacctctgac ttgagcgtcg atttttgtga tgctcgtcag gggggcggag 10500 cctatggaaa aacgccagca acgcggcc 10528 SEQ ID NO:2 pCCL-βAS3-(+mGATA-HS2-Ank-dIVS2-CoreLCRopti)-FB optimized vector.

(10483 bp)

atttttacgg ttcctggcct tttgctggcc ttttgctcac atgttctttc ctgcgrttatc 60 ccctgattct grtggataacc ςΓtattaccgc ctttgagtga gctgataccg ctcgccgcag 120 ccgaacgacc gagcgcagcg agtcagtgag cgaggaagcg gaagagcgcc caatacgcaa 180 accgcctctc cccgcgcgtt ggccgattca ttaatgcagc tggcacgaca ggtttcccga 240 ctggaaagcg ggcagtgagc gcaacgcaat taatgtgagt tagctcactc attaggcacc 300 ceaggcttta cactttatgc ttceggctcg tatgttgtgt ggaattgtga gcggataaca 360 atttcacaca ggaaacagct atgaccatga ttacgccaag cgcgcaatta accctcacta 420 aagggaacaa aagctggagc tgcaagcttg gccattgcat acgttgtatc catatcataa 480 tatgtacatt tatattggct catgtccaac attaccgcca tgttgacatt gattattgac 540 tagttattaa tagtaatcaa ttacggggtc attagttcat agcccatata tggagttccg 600 cgttacataa cttacggtaa atggcccgcc tggctgaccg cccaacgacc cccgcccatt 660 gacgtcaata atgacgtatg ttcccatagt aacgccaata gggactttcc attgacgtca 720 atgggtggag tatttacggt aaactgccca cttggcagta catcaagtgt atcatatgcc 780 aagtacgccc cctattgacg tcaatgacgg taaatggccc gcctggcatt atgcccagta 840 catgacctta tgggactttc ctacttggca gtacatctac gtattagtca tcgctattac 900 catggtgatg cggttttggc agtacatcaa tgggcgtgga tagcggtttg actcacgggg 960 atttccaagt ctccacecca ttgacgtcaa tgggagtttg ttttggcacc aaaatcaacg 1020 ggactttcca aaatgtcgta acaactccgc cccattgacg caaatgggcg gtaggcgtgt 1080 acggtgggag gtctatataa gcagagctcg tttagtgaac cggggtctct ctggttagac 1140 cagatctgag cctgggagct ctctggctaa ctagggaacc cactgcttaa gcctcaataa 1200 agcttgcctt gagtgcttca agtagtgtgt gcccgtctgt tgtgtgactc tggtaactag 1260 agatccctca gaccctttta gtcagtgtgg aaaatctcta gcagtggcgc ccgaacaggg 1320 acttgaaagc gaaagggaaa ccagaggagc tctctcgacg caggactcgg cttgctgaag 1380 cgcgcacggc aagaggcgag gggcggcgac tggtgagtac gccaaaaatt ttgactagcg 1440 gaggctagaa ggagagagat gggtgcgaga gcgtcagtat taagcggggg agaattagat 1500 cgcgatggga aaaaattcgg ttaaggccag ggggaaagaa aaaatataaa ttaaaacata 1560 tagtatgggc aagcagggag ctagaacgat tcgcagttaa tcctggcctg ttagaaacat 1620 cagaaggctg tagacaaata ctgggacagc tacaaccatc ccttcagaca ggatcagaag 1680 aacttagatc attatataat acagtagcaa ccctctattg tgtgcatcaa aggatagaga 1740 taaaagacac caaggaagct ttagacaaga tagaggaaga gcaaaacaaa agtaagacca 1800 ccgcacagca agcggccgct gatcttcaga cctggaggag gagatatgag ggacaattgg 1860 agaagtgaat tatataaata taaagtagta aaaattgaac cattaggagt agcacccacc 1920 aaggcaaaga gaagagtggt gcagagagaa aaaagagcag tgggaatagg agctttgttc 1980 cttgggttct tgggagcagc aggaagcact atgggcgcag cgtcaatgac gctgacggta 2040 caggccagac aattattgtc tggtatagtg cagcagcaga acaatttgct gagggctatt 2100 gaggcgcaae agcatctgtt gcaactcaca gtctggggca tcaagcagct ccaggcaaga 2160 atcctggctg tggaaagata cctaaaggat caacagctcc tggggatttg gggttgctct 2220 ggaaaactca tttgcaccac tgctgtgcct tggaatgcta gttggagtaa taaatctctg 2280 gaacagattt ggaatcacac gacctggatg gagtgggaca gagaaattaa caattacaca 2340 agcttaatac actccttaat tgaagaatcg caaaaccagc aagaaaagaa tgaacaagaa 2400 ttattggaat tagataaatg ggcaagtttg tggaattggt ttaacataac aaattggctg 2460 tggtatataa aattattcat aatgatagta ggaggcttgg taggtttaag aatagttttt 2520 gctgtacttt ctatagtgaa tagagttagg cagggatatt caccattatc gtttcagacc 2580 cacctcccaa ccccgagggg acccgacagg cccgaaggaa tagaagaaga aggtggagag 2640 agagacagag acagatccat tcgattagtg aacggatctc gacggtatcg atctcgacac 2700 aaatggcagt attcatccac aattttaaaa gaaaaggggg gattgggggg tacagtgcag 2760 gggaaagaat agtagacata atagcaacag acatacaaac taaagaatta caaaaacaaa 2820 ttacaaaaat tcaaaatttt cgggtttatt acagggacag cagagatcca gtttggg Leg 2880 aggatatcgg atcggaattc tctagatgat caggatccct cgagccctta tcgatcacga 2940 gactagcctc gactactagt ggagatcccc cgggctgcag agccagaagc accataaggg 3000 acatgataag ggagccagca gacctctgat ctcttcctga atgctaatct taaacatcct 3060 gaggaagaat gggacttcca tttggggtgg gcctatgata gggtaataag acagtagtga 3120 atatcaagct acaaaaagcc ccctttcaaa ttcttctcag tcctaacttt tcatactaag 3180 cccagtcctt ccaaagcaga ctgtgaaaga gtgatagttc cgggagacta gcactgcaga 3240 ttccgggtca ctgtgagtgg gggaggcagg gaagaagggc tcacaggaca gtcaaaccat 3300 gccccctgtt tttccttctt caagtagacc tctataagac aacagagaca actaaggctg 3360 agtggccagg cgaggagaaa ccatctcgcc gtaaaacatg gaaggaacac ttcaggggaa 3420 aggtggtatc tctaagcaag agaactgagt ggagtcaagg ctgagagatg caggataagc 3480 aaatgggtag tgaaaagaca ttcatgagga cagctaaaac aataagtaat gtaaaataca 3540 gcatagcaaa actttaacct ccaaatcaag cctctacttg aatccttttc tgagggatga 3600 ataaggcata ggcatcaggg gctgttgcca atgtgcatta gctgtttgca gcctcacctt 3660 ctttcatgga grtttaagata tagtgtattt tcccaaggtt tgaactagct cttcatttct 3720 ttatgtttta aatgcactga cctcccacat tcccttttta gtaaaatatt cagaaataat 3780 ttaaatacat cattgcaatg aaaataaatg ttttttatta ggcagaatcc agatgctcaa 3840 ggcccttcat aatatccccc agtttagtag ttggacttag ggaacaaagg aacctttaat 3900 agaaattgga cagcaagaaa gcgagcttag tgatacttgt gggccagggc attagccaca 3960 ccagccacca ctttctgata ggcagcctgc actggtgggg tgaattcttt gccaaagtga 4020 tgggccagca cacagaccag cacgttgccc aggagctgtg ggaggaagat aagaggtatg 4080 aacatgatta gcaaaagggc ctagcttgga ctcagaataa tccagcctta tcccaaccat 4140 aaaataaaag cagaatggta gctggattgt agctgctatt agcaatatga aacctcttac 4200 atcagttaca atttatatgc agaaataccc tgttacttct ccccttccta tgacatgaac 4260 ttaaccatag aaaagaaggg gaaagaaaac atcaagggtc ccatagactc accctgaagt 4320 tctcaggatc cacgtgcagc ttgtcacagt gcagctcact cagctgggca aaggtgccct 4380 tgaggttgtc caggtgagcc aggccatcac taaaggcacc gagcactttc ttgccatgag 4440 ccttcacctt agggttgccc ataacagcat caggagtgga cagatcccca aaggactcaa 4500 agaacctctg ggtccaaggg tagaccacca gcagcctaag ggtgggaaaa tagaccaata 4560 ggcagagaga gtcagtgcct atcagaaacc caagagtctt ctctgtctcc acatgcccag 4620 tttctattgg tctccttaaa cctgtcttgt aaccttgata ccaacctgcc cagggcctca 4680 ccaccaacgg catccacgtt caccttgtcc cacagggcag taacggcaga cttctcctca 4740 ggagtcaggt gcaccatggt gtctgtttga ggttgctagt gaacacagtt gtgtcagaag 4800 caaatgtaag caatagatgg ctctgccctg acttttatgc ccagccctgg ctcctgccct 4860 ccctgctcct gggagtagat tggccaaecc tagggtgtgg ctccacaggg tgaggtctaa 4920 gtgatgacag ccgtacctgt ccttggctct tctggcactg gcttaggagt tggacttcaa 4980 accctcagcc ctccctctaa gatatatctc ttggccccat accatcagta caaattgcta 5040 ctaaaaacat cctcctttgc aagtgtattt acccgatacg tatatgtgta tatatatata 5100 tatattcagg aaataatata ttctagaata tgtcacattc tgtctcaggc atccattttc 5160 tttatgatge cgtttgaggt ggagttttag tcaggtggtc agcttctcct tttttttgcc 5220 atctgccctg taagcatcct gctggggacc cagataggag tcatcactct aggctgagaa 5280 catctgggca cacaccctaa gcctcagcat gactcatcat gactcagcat tgctgtgctt 5340 gagccagaag gtttgcttag aaggttacac agaaccagaa ggcgggggtg gggcactgac 5400 cccgacaggg gcctggccag aactgctcat gcttggacta tgggaggtca ctaatggaga 5460 cacacagaaa tgtaacagga actaaggaaa aactgaagct ttgggggtat aggggagcag 5520 tcccatgtag tagtagaatg aaaaatgctg ctatgctgtg cctcccccac ctttcccatg 5580 tctgccctct actcatggtc tatctctcct ggctcctggg agtcatggac tccacccagc 5640 accaccaacc tgacctaacc acctatctga gcctgccagc ctataaccca tctgggccct 5700 gatagctggt ggccagccct gaccccaccc caccctccct ggaacctctg atagacacat 5760 ctggcacacc agctcgcaaa gtcaccgtga gggtcttgtg tttgctgagt caaaattcct 5820 tgaaatccaa gtccttagag actcccaggc ttggattcaa agctcctgac tttctgtcta 5880 gtgtatgtgc agtgagcccc ttttcctcta actgaaagaa ggaaaaaaaa atggaaccca 5940 aaatattcta catagtttcc atgtcacagc cagggctggg cagtctcctg ttatttcttt 6000 taaaataaat atatcattta aatgcataaa taagcaaacc ctgctcggga atgggaggga 6060 gagtctctgg agtccacccc ttctcggccc tggctctgca gatagtgcta tcaaagccct 6120 gacagagccc tgcccattgc tgggccttgg agtgagtcag cctagtagag aggcagggca 6180 agccatctca tagctgctga gtgggagaga gaaaagggct cattgtctat aaactcaggt 6240 catggctatt cttatcctgt ccctcctttt catgtaccat atttctcttc ctctttctgt 6300 gtctcctctt tccttcctcc tttactttcc ttctaacctt cctctttctc ctcctccggc 6360 aagcctttgc ttctctttct cccattcttc aaggcctcct ccatttcctc tttttattct 6420 ctcttcccct tcctttcttt ccttctgcag aggcagagac gtgcgggcca ggcccccgag 6480 ggccttaacg gccccagagg cgcttgctgt cgggccgggc gctcccggca cgggcgggcg 6540 gaggggtggc gcccgcct-gg ggaccgcaga ttacaagagc acctcctccc ccaaccccag 6600 gaggccccgc tccccatggc ctactcgacc acgagggaat tccgataatc aacctctgga 6660 ttacaaaatt tgtgaaagat tgactggtat tcttaactat gttgctcctt ttacgctatg 6720 tggatacgct gctttaatgc ctttgtatca tgctattgct tcccgtatgg ctttcatttt 6780 ctcctccttg tataaatcct ggttgctgtc tctttatgag gagttgtggc ccgttgtcag 6840 gcaacgtggc gtggtgtgca ctgtgtttgc tgacgcaacc cccactggtt ggggcattgc 6900 caccacctgt cagctccttt ccgggacttt cgctttcccc ctccctattg ccacggcgga 6960 aetcatcgce gcctgcettg cccgctgctg gacaggggct cggctgttgg gcactgacaa 7020 ttccgtggtg ttgtcgggga aatcatcgtc ctttccttgg ctgctcgcct gtgttgccac 7080 ctggattctg cgcgggacgt ccttctgcta cgtcccttcg gccctcaatc cagcggacct 7140 tccttcccgc ggcctgctgc cggctctgcg gcctcttccg cgtcttcgcc ttcgccctca 7200 gacgagtcgg atctcccttt gggccgcctc cccgcatcga taccgtcgac ctcgagacct 7260 agaaaaacat ggccaattcg agctcggtac ctttaagacc aatgacttac aaggcagctg 7320 tagatcttag ccacttttta aaagaaaagg ggggactgga agggctaatt cactcccaac 7380 gaagacaaga tcccagggat gtacgtccct aacccgctag ggggcagcac ccaggcctgc 7440 actgccgcct gccggcaggg gtccagtcct gctttttgct tgtactgggt ctctctggtt 7500 agaccagatc tgagcctggg agctctctgg ctaactaggg aacccactgc ttaagcctca 7560 ataaagcttg ccttgagtgc ttcaagtagt gtgtgcccgt ctgttgtgtg actctggtaa 7620 ctagagatcc ctcagaccct tttagtcagt gtggaaaatc tctagcagta gtagttcatg 7680 tcatcttatt attcagtatt tataacttgc aaagaaatga atatcagaga gtgagaggaa 7740 cttgtttatt gcagcttata atggttacaa ataaagcaat agcatcacaa atttcacaaa 7800 taaagcattt ttttcactgc attctagttg tggtttgtcc aaactcatca atgtatctta 7860 tcatgtctgg ctctagctat cccgccccta actccgccca tcccgcccct aactccgccc 7920 agttccgccc attctccgcc ccatggctga ctaatttttt ttatttatgc agaggccgag 7980 gccgcctcgg cctctgagct attccagaag tagtgaggag gcttttttgg aggcctaggc 8040 ttttgcgtcg agacgtaccc aattcgccct atagtgagtc gtattacgcg cgctcactgg 8100 ccgtcgtttt acaacgtcgt gactgggaaa accctggcgt tacccaactt aatcgccttg 8160 cagcacatcc ccctttcgcc agctggcgta atagcgaaga ggcccgcacc gatcgccctt 8220 cccaacagtt gcgcagcctg aatggcgaat ggcgcgacgc gccctgtagc ggcgcattaa 8280 gcgcggcggg tgtggtggtt acgcgcagcg tgaccgctac acttgccagc gccctagcgc 8340 ccgctccttt cgctttcttc ccttcctttc tcgccacgtt cgccggcttt ccccgtcaag 8400 ctctaaatcg ggggctccct ttagggttcc gatttagtgc tttacggcac ctcgacccca 8460 aaaaacttga ttagggtgat ggttcacgta gtgggccatc gccctgatag acggtttttc 8520 gccctttgac gttggagtcc acgttcttta atagtggact cttgttccaa actggaacaa 8580 cactcaaccc tatctcggtc tattcttttg atttataagg gattttgccg atttcggcct 8640 attggttaaa aaatgagctg atttaacaaa aatttaacgc gaattttaac aaaatattaa 8700 cgtttacaat ttcccaggtg gcacttttcg gggaaatgtg cgcggaaccc ctatttgttt 8760 atttttctaa atacattcaa atatgtatcc gctcatgaga caataaccct gataaatgct 8820 tcaataatat tgaaaaagga agagtatgag tattcaacat ttccgtgtcg cccttattcc 8880 cttttttgcg gcattttgcc ttcctgtttt tgctcaccca gaaacgctgg tgaaagtaaa 8940 agatgctgaa gatcagttgg gtgeacgagt gggttacatc gaactggatc tcaacagcgg 9000 taagatcctt gagagttttc gccccgaaga acgttttcca atgatgagca cttttaaagt 9060 tctgctatgt ggcgcggtat tatcccgtat tgacgccggg caagagcaac tcggtcgccg 9120 catacactat tctcagaatg acttggttga gtactcacca gtcacagaaa agcatcttac 9180 ggatggcatg acagtaagag aattatgcag tgctgccata accatgagtg ataacactgc 9240 ggccaactta cttctgacaa cgatcggagg accgaaggag ctaaccgctt ttttgcacaa 9300 catgggggat catgtaactc gccttgatcg ttgggaaccg gagctgaatg aagccatacc 9360 aaacgacgag cgtgacacca cgatgcctgt agcaatggca acaacgttgc gcaaactatt 9420 aactggcgaa ctacttactc tagcttcccg gcaacaatta atagactgga tggaggcgga 9480 taaagttgca ggaccacttc tgcgctcggc ccttccggct ggctggttta ttgctgataa 9540 atctggagcc ggtgagcgtg ggtctcgcgg tatcattgca gcactggggc cagatggtaa 9600 gccctcccgt atcgtagtta tctacacgac ggggagtcag gcaactatgg atgaacgaaa 9660 tagacagatc gctgagatag gtgcctcact gattaagcat tggtaactgt cagaccaagt 9720 ttactcatat atactttaga ttgatttaaa acttcatttt taatttaaaa ggatctaggt 9780 gaagatcctt tttgataatc tcatgaccaa aatcccttaa cgtgagtttt cgttccactg 9840 agcgtcagac cccgtagaaa agatcaaagg atcttcttga gatccttttt ttctgcgcgt 9900 aatctgctgc ttgcaaacaa aaaaaccacc gctaccagcg gtggtttgtt tgccggatca 9960 agagctacca actctttttc cgaaggtaac tggcttcagc agagcgcaga taccaaatac 10020 tgtccttcta gtgtagccgt agttaggcca ccacttcaag aactctgtag caccgcctac 10080 atacctcgct ctgctaatcc tgttaccagt ggctgctgcc agtggcgata agtcgtgtct 10140 taccgggttg gactcaagac gatagttacc ggataaggcg cagcggtcgg gctgaacggg 10200 gggttcgtgc acacagccca gcttggagcg aacgacctac accgaactga gatacctaca 10260 gcgtgagcta tgagaaagcg ccacgcttcc cgaagggaga aaggcggaca ggtatccggt 10320 aagcggcagg gtcggaacag gagagcgcac gagggagcfrb ccagggggaa acgcctggta 10380 tctttatagt cctgtcgggt ttcgccacct ctgacttgag cgtcgatttt tgtgatgctc 10440 gtcagggggg cggagcctat ggaaaaacgc cagcaacgcg gcc 10483